1
|
Tanaka S, Hamada Y, Yokoyama Y, Yamamoto H, Kogo M. Osteopontin-derived synthetic peptide SVVYGLR upregulates functional regeneration of oral and maxillofacial soft-tissue injury. JAPANESE DENTAL SCIENCE REVIEW 2021; 57:174-181. [PMID: 34630775 PMCID: PMC8487951 DOI: 10.1016/j.jdsr.2021.09.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2021] [Revised: 08/31/2021] [Accepted: 09/13/2021] [Indexed: 01/03/2023] Open
Abstract
Wound healing in the oral and maxillofacial region is a complicated and interactive process. Severe mucosal or skeletal muscle injury by trauma or surgery induces worse healing conditions, including delayed wound closure and repair with excessive scar tissue. These complications lead to persistent functional impairment, such as digestive behavior or suppression of maxillofacial growth in infancy. Osteopontin (OPN), expressed in a variety of cells, is multifunctional and comprises a number of functional domains. Seven amino acids sequence, SVVYGLR (SV peptide), exposed by thrombin cleavage of OPN, has angiogenic activity and promotes fibroblast differentiation into myofibroblasts and increased expression of collagen type III. Additionally, synthetic SV peptide shows faster dermal and oral mucosal wound closure by facilitating cell motility and migratory activities in dermal- or mucosal-derived keratinocytes and fibroblasts. Moreover, cell motility and differentiation in myogenic cell populations are accelerated by SV peptide, which contributes to the facilitation of matured myofibers and scarless healing and favorable functional regeneration after skeletal muscle injury. SV peptide has high affinity with TGF-β, with potential involvement of the TGF-β/Smad signaling pathway. Clinical application of single-dose SV peptide could be a powerful alternative treatment option for excessive oral and maxillofacial wound care to prevent disadvantageous events.
Collapse
Affiliation(s)
- Susumu Tanaka
- The 1st Department of Oral and Maxillofacial Surgery, Graduate School of Dentistry, Osaka University, Osaka, Japan
| | - Yoshinosuke Hamada
- Department of Molecular Pathology, Division of Health Sciences, Graduate School of Medicine, Osaka University, Osaka, Japan
- Department of Health Economics and Management, Graduate School of Medicine, Osaka University, Osaka, Japan
- Department of Pediatric Dentistry, Osaka Dental University, Osaka, Japan
| | - Yuhki Yokoyama
- Department of Molecular Pathology, Division of Health Sciences, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Hirofumi Yamamoto
- Department of Molecular Pathology, Division of Health Sciences, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Mikihiko Kogo
- The 1st Department of Oral and Maxillofacial Surgery, Graduate School of Dentistry, Osaka University, Osaka, Japan
| |
Collapse
|
2
|
Hou H, Wang J, Wang J, Tang W, Shaikh AS, Li Y, Fu J, Lu L, Wang F, Sun F, Tan H. A Review of Bioactive Peptides: Chemical Modification, Structural Characterization and Therapeutic Applications. J Biomed Nanotechnol 2021; 16:1687-1718. [PMID: 33485398 DOI: 10.1166/jbn.2020.3001] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
In recent years, the development and applications of protein drugs have attracted extensive attention from researchers. However, the shortcomings of protein drugs also limit their further development. Therefore, bioactive peptides isolated or simulated from protein polymers have broad application prospects in food, medicine, biotechnology, and other industries. Such peptides have a molecular weight distribution between 180 and 1000 Da. As a small molecule substance, bioactive peptide is usually degraded by various enzymes in the organism and have a short half-life. At the same time, such substances have poor stability and are difficult to produce and store. Therefore, these active peptides may be modified through phosphorylation, glycosylation, and acylation. Compared with other protein drugs, the modified active peptides are more easily absorbed by the body, have longer half-life, stronger targeting, and fewer side effects in addition to higher bioavailability. In the light of their functions, bioactive peptide can be divided into antimicrobial, anti-tumour, anti-angiogenic, antioxidant, anti-fatigue, and anti-hypertensive peptides. This article mainly focuses on the introduction of several promising biologically active peptides functioning as antimicrobial, anti-tumour, antiangiogenic, and antioxidant peptides from the three aspects modification, structural characteristics and mechanism of action.
Collapse
|
3
|
Jendryczko K, Chudzian J, Skinder N, Opaliński Ł, Rzeszótko J, Wiedlocha A, Otlewski J, Szlachcic A. FGF2-Derived PeptibodyF2-MMAE Conjugate for Targeted Delivery of Cytotoxic Drugs into Cancer Cells Overexpressing FGFR1. Cancers (Basel) 2020; 12:E2992. [PMID: 33076489 PMCID: PMC7602595 DOI: 10.3390/cancers12102992] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2020] [Accepted: 10/10/2020] [Indexed: 12/23/2022] Open
Abstract
Fibroblast growth factor receptors (FGFRs) are emerging targets for directed cancer therapy. Presented here is a new FGFR1-targeting conjugate, the peptibodyF2, which employs peptibody, a fusion of peptide and the Fc fragment of human IgG as a selective targeting agent and drug carrier. Short peptide based on FGF2 sequence was used to construct a FGFR1-targeting peptibody. We have shown that this peptide ensures specific delivery of peptibodyF2 into FGFR1-expressing cells. In order to use peptibodyF2 as a delivery vehicle for cytotoxic drugs, we have conjugated it with MMAE, a drug widely used in antibody-drug conjugates for targeted therapy. Resulting conjugate shows high and specific cytotoxicity towards FGFR1-positive cells, i.e., squamous cell lung carcinoma NCI-H520, while remaining non-toxic for FGFR1-negative cells. Such peptibody-drug conjugate can serve as a basis for development of therapy for tumors with overexpressed or malfunctioning FGFRs.
Collapse
Affiliation(s)
- Karolina Jendryczko
- Department of Protein Engineering, Faculty of Biotechnology, University of Wroclaw, 50383 Wroclaw, Poland; (K.J.); (J.C.); (N.S.); (Ł.O.); (J.R.); (J.O.)
| | - Julia Chudzian
- Department of Protein Engineering, Faculty of Biotechnology, University of Wroclaw, 50383 Wroclaw, Poland; (K.J.); (J.C.); (N.S.); (Ł.O.); (J.R.); (J.O.)
| | - Natalia Skinder
- Department of Protein Engineering, Faculty of Biotechnology, University of Wroclaw, 50383 Wroclaw, Poland; (K.J.); (J.C.); (N.S.); (Ł.O.); (J.R.); (J.O.)
| | - Łukasz Opaliński
- Department of Protein Engineering, Faculty of Biotechnology, University of Wroclaw, 50383 Wroclaw, Poland; (K.J.); (J.C.); (N.S.); (Ł.O.); (J.R.); (J.O.)
| | - Jakub Rzeszótko
- Department of Protein Engineering, Faculty of Biotechnology, University of Wroclaw, 50383 Wroclaw, Poland; (K.J.); (J.C.); (N.S.); (Ł.O.); (J.R.); (J.O.)
| | - Antoni Wiedlocha
- Department of Molecular Cell Biology, Institute for Cancer Research, The Norwegian Radium Hospital, Oslo University Hospital, 0379 Oslo, Norway;
- Department of Radiobiology and Radiation Protection, Military Institute of Hygiene and Epidemiology, 01163 Warsaw, Poland
- Center for Cancer Cell Reprogramming, Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, 0379 Oslo, Norway
| | - Jacek Otlewski
- Department of Protein Engineering, Faculty of Biotechnology, University of Wroclaw, 50383 Wroclaw, Poland; (K.J.); (J.C.); (N.S.); (Ł.O.); (J.R.); (J.O.)
| | - Anna Szlachcic
- Department of Protein Engineering, Faculty of Biotechnology, University of Wroclaw, 50383 Wroclaw, Poland; (K.J.); (J.C.); (N.S.); (Ł.O.); (J.R.); (J.O.)
| |
Collapse
|
4
|
Lian Z, Du W, Zhang Y, Fu Y, Liu T, Wang A, Cai T, Zhu J, Zeng Y, Liu Z, Huang JA. Anlotinib can overcome acquired resistance to EGFR-TKIs via FGFR1 signaling in non-small cell lung cancer without harboring EGFR T790M mutation. Thorac Cancer 2020; 11:1934-1943. [PMID: 32433828 PMCID: PMC7327692 DOI: 10.1111/1759-7714.13485] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2020] [Revised: 04/27/2020] [Accepted: 04/28/2020] [Indexed: 12/24/2022] Open
Abstract
Background Although many studies have defined mechanisms of resistance to EGFR‐TKIs, acquired resistance remains the major limitation of monotherapy with EGFR‐TKIs. Methods Cell viability was analyzed using a Cell Counting Kit‐8 (CCK‐8) assay. EGFR T790M mutation was sequenced on a HiSeq 4000 platform. mRNAs from HCC827 and HCC827 gefitinib‐resistant (GR) cells were analyzed by genome analyzer‐based deep sequencing. The effect of anlotinib on apoptosis and cell cycle arrest of HCC827 GR was detected by fluorescence‐activated cell sorting (FACS) analysis. A mouse xenograft model was used to assess the effect of anlotinib on HCC827 GR cells. Results The T790M mutation was found in the PC‐9 GR cell line but not in the HCC827 GR cell line. Anlotinib could suppress the growth of HCC827 GR cells by inhibiting FGFR1 in vitro and in a mouse xenograft model. Moreover, FGFR1 was overexpressed in HCC827 GR cells, and the knockdown of FGFR1 reversed gefitinib resistance in HCC827 GR cells. Furthermore, anlotinib induced apoptosis and cell cycle arrest in HCC827 GR cells by increasing the activity of Caspase‐3. Conclusions FGFR1 overexpression could be the mechanism of EGFR‐TKI acquired resistance and anlotinib can suppresse the growth of EGFR‐TKI‐resistant NSCLC cells without T790M mutation.
Collapse
Affiliation(s)
- Zengzhi Lian
- Department of Respiratory Medicine, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Wenwen Du
- Department of Respiratory Medicine, The First Affiliated Hospital of Soochow University, Suzhou, China.,Institute of Respiratory Diseases, Soochow University, Suzhou, China
| | - Yang Zhang
- Department of Respiratory Medicine, The First Affiliated Hospital of Soochow University, Suzhou, China.,Institute of Respiratory Diseases, Soochow University, Suzhou, China
| | - Yulong Fu
- Department of Respiratory Medicine, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Ting Liu
- Department of Respiratory Medicine, The First Affiliated Hospital of Soochow University, Suzhou, China.,Institute of Respiratory Diseases, Soochow University, Suzhou, China
| | - Anqi Wang
- Department of Respiratory Medicine, The First Affiliated Hospital of Soochow University, Suzhou, China.,Institute of Respiratory Diseases, Soochow University, Suzhou, China
| | - Tingting Cai
- Department of Respiratory Medicine, The First Affiliated Hospital of Soochow University, Suzhou, China.,Institute of Respiratory Diseases, Soochow University, Suzhou, China
| | - Jianjie Zhu
- Department of Respiratory Medicine, The First Affiliated Hospital of Soochow University, Suzhou, China.,Institute of Respiratory Diseases, Soochow University, Suzhou, China.,Suzhou Key Laboratory for Respiratory Diseases, Suzhou, China
| | - Yuanyuan Zeng
- Department of Respiratory Medicine, The First Affiliated Hospital of Soochow University, Suzhou, China.,Institute of Respiratory Diseases, Soochow University, Suzhou, China.,Suzhou Key Laboratory for Respiratory Diseases, Suzhou, China
| | - Zeyi Liu
- Department of Respiratory Medicine, The First Affiliated Hospital of Soochow University, Suzhou, China.,Institute of Respiratory Diseases, Soochow University, Suzhou, China.,Suzhou Key Laboratory for Respiratory Diseases, Suzhou, China
| | - Jian-An Huang
- Department of Respiratory Medicine, The First Affiliated Hospital of Soochow University, Suzhou, China.,Institute of Respiratory Diseases, Soochow University, Suzhou, China.,Suzhou Key Laboratory for Respiratory Diseases, Suzhou, China
| |
Collapse
|
5
|
Dianat-Moghadam H, Teimoori-Toolabi L. Implications of Fibroblast Growth Factors (FGFs) in Cancer: From Prognostic to Therapeutic Applications. Curr Drug Targets 2019; 20:852-870. [DOI: 10.2174/1389450120666190112145409] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2018] [Revised: 01/01/2019] [Accepted: 01/02/2019] [Indexed: 12/22/2022]
Abstract
Fibroblast growth factors (FGFs) are pleiotropic molecules exerting autocrine, intracrine
and paracrine functions via activating four tyrosine kinase FGF receptors (FGFR), which further trigger
a variety of cellular processes including angiogenesis, evasion from apoptosis, bone formation,
embryogenesis, wound repair and homeostasis. Four major mechanisms including angiogenesis, inflammation,
cell proliferation, and metastasis are active in FGF/FGFR-driven tumors. Furthermore,
gain-of-function or loss-of-function in FGFRs1-4 which is due to amplification, fusions, mutations,
and changes in tumor–stromal cells interactions, is associated with the development and progression
of cancer. Although, the developed small molecule or antibodies targeting FGFR signaling offer immense
potential for cancer therapy, emergence of drug resistance, activation of compensatory pathways
and systemic toxicity of modulators are bottlenecks in clinical application of anti-FGFRs. In this
review, we present FGF/FGFR structure and the mechanisms of its function, as well as cross-talks
with other nodes and/or signaling pathways. We describe deregulation of FGF/FGFR-related mechanisms
in human disease and tumor progression leading to the presentation of emerging therapeutic approaches,
resistance to FGFR targeting, and clinical potentials of individual FGF family in several
human cancers. Additionally, the underlying biological mechanisms of FGF/FGFR signaling, besides
several attempts to develop predictive biomarkers and combination therapies for different cancers
have been explored.
Collapse
Affiliation(s)
- Hassan Dianat-Moghadam
- Molecular Medicine Department, Biotechnology Research Center, Pasteur Institute of Iran, Tehran, Iran
| | - Ladan Teimoori-Toolabi
- Molecular Medicine Department, Biotechnology Research Center, Pasteur Institute of Iran, Tehran, Iran
| |
Collapse
|
6
|
Gammelgaard KR, Vad-Nielsen J, Clement MS, Weiss S, Daugaard TF, Dagnæs-Hansen F, Meldgaard P, Sorensen BS, Nielsen AL. Up-Regulated FGFR1 Expression as a Mediator of Intrinsic TKI Resistance in EGFR-Mutated NSCLC. Transl Oncol 2019; 12:432-440. [PMID: 30562682 PMCID: PMC6297127 DOI: 10.1016/j.tranon.2018.11.017] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2018] [Revised: 11/27/2018] [Accepted: 11/27/2018] [Indexed: 12/20/2022] Open
Abstract
Non-small cell lung carcinoma patients with epidermal growth factor receptor (EGFR) mutations are offered EGFR tyrosine kinase inhibitors (TKI) as first line treatment, but 20-40% of these patients do not respond. High expression of alternative receptor tyrosine kinases, such as Fibroblast growth factor receptor 1 (FGFR1), potentially mediates intrinsic EGFR TKI resistance. To study this in molecular detail, we used CRISPR-dCas9 Synergistic Activation Mediator (SAM) for up-regulation of FGFR1 in physiological relevant levels in the EGFR mutated NSCLC cell lines HCC827 and PC9 thereby generating HCC827gFGFR1 and PC9gFGFR1. The sensitivity to the TKI erlotinib was investigated in vitro and in a BALBc nu/nu mouse xenograft model. FGFR1 up-regulation decreased TKI-sensitivity in both NSCLC cell lines in the presence of the ligand fibroblast growth factor 2 (FGF2). Xenografts were established with PC9gFGFR1 cells and it was demonstrated that there was no significant difference in tumor size between TKI- and vehicle-treated PC9gFGFR1 tumors. This supports decreased TKI-sensitivity in NSCLC cells with FGFR1 up-regulation. Our study points to FGFR1 signaling being an intrinsic resistance mechanism abolishing TKI response in EGFR mutated NSCLC.
Collapse
Affiliation(s)
- Kristine Raaby Gammelgaard
- Department of Biomedicine, Aarhus University, Aarhus, Denmark; Department of Clinical Biochemistry, Aarhus University Hospital, Aarhus, Denmark
| | | | | | - Simone Weiss
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | | | | | - Peter Meldgaard
- Department of Oncology, Aarhus University Hospital, Aarhus, Denmark
| | - Boe Sandahl Sorensen
- Department of Clinical Biochemistry, Aarhus University Hospital, Aarhus, Denmark
| | | |
Collapse
|
7
|
D'Arcangelo D, Facchiano F, Nassa G, Stancato A, Antonini A, Rossi S, Senatore C, Cordella M, Tabolacci C, Salvati A, Tarallo R, Weisz A, Facchiano AM, Facchiano A. PDGFR-alpha inhibits melanoma growth via CXCL10/IP-10: a multi-omics approach. Oncotarget 2018; 7:77257-77275. [PMID: 27764787 PMCID: PMC5363585 DOI: 10.18632/oncotarget.12629] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2016] [Accepted: 09/25/2016] [Indexed: 12/21/2022] Open
Abstract
Melanoma is the most aggressive skin-cancer, showing high mortality at advanced stages. Platelet Derived Growth Factor Receptor-alpha (PDGFR-alpha) potently inhibits melanoma- and endothelium-proliferation and its expression is significantly reduced in melanoma-biopsies, suggesting that melanoma progression eliminates cells expressing PDGFR-alpha. In the present study transient overexpression of PDGFR-alpha in endothelial (HUVEC) and melanoma (SKMel-28, A375, Preyer) human-cells shows strong anti-proliferative effects, with profound transcriptome and miRNome deregulation. PDGFR-alpha overexpression strongly affects expression of 82 genes in HUVEC (41 up-, 41 down-regulated), and 52 genes in SKMel-28 (43 up-, 9 down-regulated). CXCL10/IP-10 transcript showed up to 20 fold-increase, with similar changes detectable at the protein level. miRNA expression profiling in cells overexpressing PDGFR-alpha identified 14 miRNAs up- and 40 down-regulated, with miR-503 being the most down-regulated (6.4 fold-reduction). miR-503, miR-630 and miR-424 deregulation was confirmed by qRT-PCR. Interestingly, the most upregulated transcript (i.e., CXCL10/IP-10) was a validated miR-503 target and CXCL10/IP-10 neutralization significantly reverted the anti-proliferative action of PDGFR-alpha, and PDGFR-alpha inhibition by Dasatinb totally reverted the CXCL10/IP10 induction, further supporting a functional interplay of these factors. Finally, integration of transcriptomics and miRNomics data highlighted several pathways affected by PDGFR-alpha. This study demonstrates for the first time that PDGFR-alpha strongly inhibits endothelial and melanoma cells proliferation in a CXCL10/IP-10 dependent way, via miR-503 down-regulation.
Collapse
Affiliation(s)
- Daniela D'Arcangelo
- Istituto Dermopatico dell'Immacolata, IDI-IRCCS, Fondazione Luigi Maria Monti, Rome, Italy
| | - Francesco Facchiano
- Dipartimento Ematologia, Oncologia e Medicina Molecolare, Istituto Superiore di Sanità, Rome, Italy
| | - Giovanni Nassa
- Laboratory of Molecular Medicine and Genomics, Department of Medicine and Surgery,University of Salerno, Baronissi (SA), Italy.,Genomix4Life srl, Department of Medicine and Surgery, University of Salerno, Baronissi (SA), Italy
| | - Andrea Stancato
- Istituto Dermopatico dell'Immacolata, IDI-IRCCS, Fondazione Luigi Maria Monti, Rome, Italy
| | - Annalisa Antonini
- Istituto Dermopatico dell'Immacolata, IDI-IRCCS, Fondazione Luigi Maria Monti, Rome, Italy
| | - Stefania Rossi
- Dipartimento Ematologia, Oncologia e Medicina Molecolare, Istituto Superiore di Sanità, Rome, Italy
| | - Cinzia Senatore
- Dipartimento Ematologia, Oncologia e Medicina Molecolare, Istituto Superiore di Sanità, Rome, Italy
| | - Martina Cordella
- Dipartimento Ematologia, Oncologia e Medicina Molecolare, Istituto Superiore di Sanità, Rome, Italy
| | - Claudio Tabolacci
- Dipartimento Ematologia, Oncologia e Medicina Molecolare, Istituto Superiore di Sanità, Rome, Italy
| | - Annamaria Salvati
- Laboratory of Molecular Medicine and Genomics, Department of Medicine and Surgery,University of Salerno, Baronissi (SA), Italy
| | - Roberta Tarallo
- Laboratory of Molecular Medicine and Genomics, Department of Medicine and Surgery,University of Salerno, Baronissi (SA), Italy
| | - Alessandro Weisz
- Laboratory of Molecular Medicine and Genomics, Department of Medicine and Surgery,University of Salerno, Baronissi (SA), Italy
| | | | - Antonio Facchiano
- Istituto Dermopatico dell'Immacolata, IDI-IRCCS, Fondazione Luigi Maria Monti, Rome, Italy
| |
Collapse
|
8
|
Brouwers B, Fumagalli D, Brohee S, Hatse S, Govaere O, Floris G, Van den Eynde K, Bareche Y, Schöffski P, Smeets A, Neven P, Lambrechts D, Sotiriou C, Wildiers H. The footprint of the ageing stroma in older patients with breast cancer. Breast Cancer Res 2017; 19:78. [PMID: 28673354 PMCID: PMC5494807 DOI: 10.1186/s13058-017-0871-0] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2016] [Accepted: 06/20/2017] [Indexed: 12/14/2022] Open
Abstract
Background Tumours are not only composed of malignant cells but also consist of a stromal micro-environment, which has been shown to influence cancer cell behaviour. Because the ageing process induces accumulation of senescent cells in the body, this micro-environment is thought to be different in cancers occurring in old patients compared with younger patients. More specifically, senescence-related fibroblastic features, such as the senescence-associated secretory profile (SASP) and the induction of autophagy, are suspected to stimulate tumour growth and progression. Methods We compared gene expression profiles in stromal fields of breast carcinomas by performing laser capture microdissection of the cancer-associated stroma from eight old (aged ≥80 years at diagnosis) and nine young (aged <45 years at diagnosis) patients with triple-negative breast cancer. Gene expression data were obtained by microarray analysis (Affymetrix). Differential gene expression and gene set enrichment analysis (GSEA) were performed. Results Differential gene expression analysis showed changes reminiscent of increased growth, de-differentiation and migration in stromal samples of older versus younger patients. GSEA confirmed the presence of a SASP, as well as the presence of autophagy in the stroma of older patients. Conclusions We provide the first evidence in humans that older age at diagnosis is associated with a different stromal micro-environment in breast cancers. The SASP and the presence of autophagy appear to be important age-induced stromal features. Electronic supplementary material The online version of this article (doi:10.1186/s13058-017-0871-0) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Barbara Brouwers
- Laboratory of Experimental Oncology (LEO), Department of Oncology, KU Leuven, Leuven, Belgium. .,Department of General Medical Oncology, Leuven Cancer Institute, University Hospitals Leuven, Leuven, Belgium.
| | - Debora Fumagalli
- Breast Cancer Translational Research Laboratory, Institut Jules Bordet, Universite Libre de Bruxelles, Brussels, Belgium
| | - Sylvain Brohee
- Breast Cancer Translational Research Laboratory, Institut Jules Bordet, Universite Libre de Bruxelles, Brussels, Belgium
| | - Sigrid Hatse
- Laboratory of Experimental Oncology (LEO), Department of Oncology, KU Leuven, Leuven, Belgium.,Department of General Medical Oncology, Leuven Cancer Institute, University Hospitals Leuven, Leuven, Belgium
| | - Olivier Govaere
- Department of Imaging and Pathology, Laboratory of Translational Cell & Tissue Research, KU Leuven, Herestraat 49, B-3000, Leuven, Belgium.,Department of Pathology, University Hospitals Leuven, Herestraat 49, B-3000, Leuven, Belgium
| | - Giuseppe Floris
- Department of Imaging and Pathology, Laboratory of Translational Cell & Tissue Research, KU Leuven, Herestraat 49, B-3000, Leuven, Belgium.,Department of Pathology, University Hospitals Leuven, Herestraat 49, B-3000, Leuven, Belgium
| | - Kathleen Van den Eynde
- Department of Imaging and Pathology, Laboratory of Translational Cell & Tissue Research, KU Leuven, Herestraat 49, B-3000, Leuven, Belgium.,Department of Pathology, University Hospitals Leuven, Herestraat 49, B-3000, Leuven, Belgium
| | - Yacine Bareche
- Breast Cancer Translational Research Laboratory, Institut Jules Bordet, Universite Libre de Bruxelles, Brussels, Belgium
| | - Patrick Schöffski
- Laboratory of Experimental Oncology (LEO), Department of Oncology, KU Leuven, Leuven, Belgium.,Department of General Medical Oncology, Leuven Cancer Institute, University Hospitals Leuven, Leuven, Belgium
| | - Ann Smeets
- Multidisciplinary Breast Center, University Hospitals Leuven, Leuven, Belgium
| | - Patrick Neven
- Multidisciplinary Breast Center, University Hospitals Leuven, Leuven, Belgium
| | - Diether Lambrechts
- Department of Oncology, Laboratory for Translational Genetics, Vesalius Research Center (VRC), Vlaams Instituut voor Biotechnologie (VIB) and KU Leuven, Leuven, Belgium
| | - Christos Sotiriou
- Breast Cancer Translational Research Laboratory, Institut Jules Bordet, Universite Libre de Bruxelles, Brussels, Belgium
| | - Hans Wildiers
- Laboratory of Experimental Oncology (LEO), Department of Oncology, KU Leuven, Leuven, Belgium.,Department of General Medical Oncology, Leuven Cancer Institute, University Hospitals Leuven, Leuven, Belgium.,Multidisciplinary Breast Center, University Hospitals Leuven, Leuven, Belgium
| |
Collapse
|
9
|
Moscaroli A, Jones G, Lühmann T, Meinel L, Wälti S, Blanc A, Fischer E, Hilbert M, Schibli R, Béhé M. Radiolabeled 111In-FGF-2 Is Suitable for In Vitro/Ex Vivo Evaluations and In Vivo Imaging. Mol Pharm 2017; 14:639-648. [PMID: 28221043 DOI: 10.1021/acs.molpharmaceut.6b00913] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Fibroblast growth factor-2 (FGF-2) is a potent modulator of cell growth and regulation, with improper FGF-2 signaling being involved in impaired responses to injury or even cancer. Therefore, the exploitation of FGF-2 as a therapeutic drives the prerequisite for effective insight into drug disposition kinetics. In this article, we present an 111In-radiolabeled FGF-2 derivative for noninvasive imaging in small animals deploying single photon emission tomography (SPECT). 111In-FGF-2 is equally well suitable for in vitro and ex vivo investigations as 125I-FGF-2. Furthermore, 111In-FGF-2 permits the performance of in vivo imaging, for example for the analysis of FGF-2 containing pharmaceutical formulations in developmental or preclinical stages. 111In-FGF-2 had affinity for the low-molecular-weight heparin enoxaparin identical to that of unlabeled FGF-2 (Kd: 0.6 ± 0.07 μM and 0.33 ± 0.03 μM, respectively) as assessed by isothermal titration calorimetry. The binding of 111In-FGF-2 to heparan sulfate proteoglycans (HPSGs) and the biological activity were comparable to those of unlabeled FGF-2, with EC50 values of 12 ± 2 pM and 25 ± 6 pM, respectively. In vivo biodistribution in healthy nude mice indicated a predominant accumulation of 111In-FGF-2 in filtering organs and minor uptake in the retina and the salivary and pituitary glands, which was confirmed by SPECT imaging. Therefore, 111In-FGF-2 is a valid tracer for future noninvasive animal imaging of FGF-2 in pharmaceutical development.
Collapse
Affiliation(s)
- Alessandra Moscaroli
- Center for Radiopharmaceutical Sciences ETH-PSI-USZ, Paul Scherrer Institute , 5232 Villigen PSI, Switzerland
| | - Gabriel Jones
- Institute for Pharmacy and Food Chemistry, University of Wurzburg , 97074 Wurzburg, Germany
| | - Tessa Lühmann
- Institute for Pharmacy and Food Chemistry, University of Wurzburg , 97074 Wurzburg, Germany
| | - Lorenz Meinel
- Institute for Pharmacy and Food Chemistry, University of Wurzburg , 97074 Wurzburg, Germany
| | - Stephanie Wälti
- Institute of Pharmaceutical Sciences, Department of Chemistry and Applied Biosciences, ETH Zurich , 8092 Zurich, Switzerland
| | - Alain Blanc
- Center for Radiopharmaceutical Sciences ETH-PSI-USZ, Paul Scherrer Institute , 5232 Villigen PSI, Switzerland
| | - Eliane Fischer
- Center for Radiopharmaceutical Sciences ETH-PSI-USZ, Paul Scherrer Institute , 5232 Villigen PSI, Switzerland
| | - Manuel Hilbert
- Laboratory of Biomolecular Research, Department of Biology and Chemistry, Paul Scherrer Institute , 5232 Villigen PSI, Switzerland
| | - Roger Schibli
- Center for Radiopharmaceutical Sciences ETH-PSI-USZ, Paul Scherrer Institute , 5232 Villigen PSI, Switzerland.,Institute of Pharmaceutical Sciences, Department of Chemistry and Applied Biosciences, ETH Zurich , 8092 Zurich, Switzerland
| | - Martin Béhé
- Center for Radiopharmaceutical Sciences ETH-PSI-USZ, Paul Scherrer Institute , 5232 Villigen PSI, Switzerland
| |
Collapse
|
10
|
Fan L, Li W, Ying S, Shi L, Wang Z, Chen G, Ye H, Wu X, Wu J, Liang G, Li X. A peptide derivative serves as a fibroblast growth factor 2 antagonist in human gastric cancer. Tumour Biol 2015; 36:7233-41. [DOI: 10.1007/s13277-015-3435-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2015] [Accepted: 04/07/2015] [Indexed: 01/26/2023] Open
|
11
|
Fan L, Xie H, Chen L, Ye H, Ying S, Wang C, Wu X, Li W, Wu J, Liang G, Li X. A novel FGF2 antagonist peptide P8 with potent antiproliferation activity. Tumour Biol 2014; 35:10571-9. [PMID: 25062723 DOI: 10.1007/s13277-014-2356-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2014] [Accepted: 07/14/2014] [Indexed: 12/23/2022] Open
Abstract
Some fibroblast growth factors (FGFs) play a critical role in tumorigenesis and progression. Among them, FGF2 was highly expressed in some tumors, and antagonists binding to FGF2 can suppress the growth of tumor cells. Therefore, FGF2 has been considered as an important target in cancer therapy. In this study, we identified a novel FGF2-binding short peptide (P8, PLLQATAGGGS-NH2) using phage display technology and alanine scanning. The P8 peptide suppressed FGF2-induced proliferation with no cytotoxic effect on cells, arrested the cycle at the G0/G1 phase in B16-F10 cells, and downregulated the activation of fibroblast growth factor receptor substrate 2α (FRS2α)/ERK cascade in B16-F10, NIH-H460, and SGC-7901 cells. Besides, P8 peptide can also inhibit the phosphorylation of FRS2α stimulated by FGF1 and KGF2. These implied that P8 peptide may develop as a multi-target antagonist peptide contributing to tumor treatment.
Collapse
Affiliation(s)
- Lei Fan
- Chemical Biology Research Center, College of Pharmaceutical Sciences, Wenzhou Medical University, 325035, Wenzhou, China
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
12
|
Ronca R, Di Salle E, Giacomini A, Leali D, Alessi P, Coltrini D, Ravelli C, Matarazzo S, Ribatti D, Vermi W, Presta M. Long Pentraxin-3 Inhibits Epithelial–Mesenchymal Transition in Melanoma Cells. Mol Cancer Ther 2013; 12:2760-71. [DOI: 10.1158/1535-7163.mct-13-0487] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
13
|
Inhibition of Proliferation of Non-small Cell Lung Cancer Cells by a bFGF Antagonist Peptide. Int J Pept Res Ther 2013. [DOI: 10.1007/s10989-013-9372-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
14
|
Yu Y, Gao S, Li Q, Wang C, Lai X, Chen X, Wang R, Di J, Li T, Wang W, Wu X. The FGF2-binding peptide P7 inhibits melanoma growth in vitro and in vivo. J Cancer Res Clin Oncol 2012; 138:1321-8. [PMID: 22481251 DOI: 10.1007/s00432-012-1201-7] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2012] [Accepted: 03/12/2012] [Indexed: 10/28/2022]
Abstract
PURPOSE Melanoma is a malignant tumor and causes majority of deaths related to skin cancer. Fibroblast growth factor 2 (FGF2) greatly contributes to melanoma growth and progress. In this paper, we attempt to evaluate the therapeutic potential of FGF2-binding peptide (named P7) using as a potent FGF2 antagonist via exploration of its antitumor effect on melanoma in vitro and in vivo. METHODS Cell viability was measured by WST-1. Cell cycle progression was determined by propidium iodide staining and flow cytometry. Western blotting was carried out to detect the activation of Erk1/2, P38, Akt, and MEK, and the expression of apoptosis-associated proteins. The influence of P7 on FGF2 internalization was assessed by separation of nuclear and cytoplasmic protein fractions followed by Western blotting. Female C57BL/6 mice bearing xenograft melanoma were established and used to evaluate the antitumor effect of P7 in vivo. RESULTS In this study, we first proved that P7 peptides significantly inhibited proliferation of FGF2-induced melanoma cell line B16-F10. Further investigations revealed that the mechanisms of P7 peptides inhibiting cell proliferation of melanoma cells stimulated with FGF2 in vitro involved cell cycle arrest at the G0/G1 phase, blockade of the activation of Erk1/2, P38, and Akt cascades, and inhibition of FGF2 internalization. Finally, treatment of P7 peptides in a murine melanoma model resulted in significant inhibition of tumor growth and angiogenesis in vivo, which was associated with blockade of mitogen-activated protein kinase signal activation, and suppression of the expressions of anti-apoptotic Bcl-2 protein and angiogenic factor in the melanoma tumors. CONCLUSIONS The FGF2-binding peptide with potent antiproliferation and anti-angiogenic activity may have therapeutic potential in melanoma.
Collapse
MESH Headings
- Amino Acid Sequence
- Animals
- Blotting, Western
- Cell Cycle Checkpoints/drug effects
- Cell Line, Tumor
- Cell Proliferation/drug effects
- Dose-Response Relationship, Drug
- Endocytosis/drug effects
- Female
- Fibroblast Growth Factor 2/antagonists & inhibitors
- Fibroblast Growth Factor 2/metabolism
- Fibroblast Growth Factor 2/pharmacology
- Flow Cytometry
- G1 Phase/genetics
- Melanoma, Experimental/metabolism
- Melanoma, Experimental/pathology
- Melanoma, Experimental/prevention & control
- Mice
- Mice, Inbred C57BL
- Mitogen-Activated Protein Kinases/metabolism
- Neovascularization, Pathologic/metabolism
- Neovascularization, Pathologic/pathology
- Neovascularization, Pathologic/prevention & control
- Oligopeptides/metabolism
- Oligopeptides/pharmacology
- Phosphorylation/drug effects
- Protein Binding
- Proto-Oncogene Proteins c-akt/metabolism
- Resting Phase, Cell Cycle/drug effects
- Tumor Burden/drug effects
Collapse
Affiliation(s)
- Yonglin Yu
- Institute of Tissue Transplantation and Immunology, Jinan University, Guangzhou 510632, China
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
15
|
Mattiello T, Guerriero R, Lotti LV, Trifirò E, Felli MP, Barbarulo A, Pucci B, Gazzaniga P, Gaudio C, Frati L, Pulcinelli FM. Aspirin extrusion from human platelets through multidrug resistance protein-4-mediated transport: evidence of a reduced drug action in patients after coronary artery bypass grafting. J Am Coll Cardiol 2011; 58:752-61. [PMID: 21816313 DOI: 10.1016/j.jacc.2011.03.049] [Citation(s) in RCA: 63] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/10/2011] [Revised: 03/18/2011] [Accepted: 03/22/2011] [Indexed: 11/19/2022]
Abstract
OBJECTIVES In this study we investigate: 1) the role of multidrug resistance protein-4 (MRP4), an organic anion unidirectional transporter, in modulating aspirin action on human platelet cyclooxygenase (COX)-1; and 2) whether the impairment of aspirin-COX-1 interaction, found in coronary artery bypass grafting (CABG) patients, could be dependent on MRP4-mediated transport. BACKGROUND Platelets of CABG patients present a reduced sensitivity to aspirin despite in vivo and in vitro drug treatment. Aspirin is an organic anion and could be a substrate for MRP4. METHODS Intracellular aspirin concentration and drug COX-1 activity, measured by thrombin-induced thromboxane B2 (TxB2) production, were evaluated in platelets obtained from healthy volunteers (HV) and hematopoietic-progenitor cell cultures reducing or not reducing MRP4-mediated transport. Platelet MRP4 expression was evaluated, in platelets from HV and CABG patients, by dot-blot or by immunogold-electromicrographs or immunofluorescence-microscopy analysis. RESULTS Inhibition of MRP4-mediated transport by dipyridamole or Mk-571 increases aspirin entrapment and its in vitro effect on COX-1 activity (142.7 ± 34.6 pg/10(8) cells vs. 343.7 ± 169.3 pg/10⁸ cells TxB2-production). Platelets derived from megakaryocytes transfected with MRP4 small interfering ribonucleic acid have a higher aspirin entrapment and drug COX-1 activity. Platelets from CABG patients showed a high expression of MRP4 whose in vitro inhibition enhanced aspirin effect on COX-1 (349 ± 141 pg/10⁸ cells vs. 1,670 ± 646 pg/10⁸ cells TxB2-production). CONCLUSIONS Aspirin is a substrate for MRP4 and can be extruded from platelet through its transportation. Aspirin effect on COX-1 is little-related to MRP4-mediated aspirin transport in HV, but in CABG patients with MRP4 over-expression, its pharmacological inhibition enhances aspirin action in an efficient way.
Collapse
Affiliation(s)
- Teresa Mattiello
- Department of Experimental Medicine, Sapienza University of Rome, Rome, Italy
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
16
|
Ong LL, Li W, Oldigs JK, Kaminski A, Gerstmayer B, Piechaczek C, Wagner W, Li RK, Ma N, Steinhoff G. Hypoxic/normoxic preconditioning increases endothelial differentiation potential of human bone marrow CD133+ cells. Tissue Eng Part C Methods 2011; 16:1069-81. [PMID: 20073989 DOI: 10.1089/ten.tec.2009.0641] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
CD133+ cells are hemangioblasts that have capacity to generate into both hematopoietic and endothelial cells (ECs). Hypoxia/normoxia has shown to be the regulator of the balance between stemness and differentiation. In this study we performed Agilent's whole human genome oligo microarray analysis and examined the differentiation potential of the bone-marrow-derived CD133+ cells after hypoxic/normoxic preconditioning of CD133+ cells. Results showed that there was no significant increase in erythroid colony forming unit (CFU-E) and CFU-granulocyte, erythrocyte, monocyte, and megakaryocyte formation with cells treated under hypoxia/normoxia. However, a significant increment of EC forming unit at 24 h (143.2 +/- 8.0%) compared to 0 h (100 +/- 11.4%) was observed in CFU-EC analysis. Reverse transcription-polymerase chain reaction and immunostaining analysis showed that the differentiated cells diminished hematopoietic stem cell surface markers and acquired the gene markers and functional phenotype of ECs. The transcriptome profile revealed a cluster of 232 downregulated and 498 upregulated genes in cells treated for 24 h under hypoxia. The upregulated genes include angiogenic genes, angiogenic growth factor genes, angiogenic cytokine and chemokine genes, as well as angiogenic-positive regulatory genes, including FGFBP1, PDGFB, CCL15, CXCL12, CXCL6, IL-6, PTN, EREG, ERBB2, EDG5, FGF3, FHF2, GDF15, JUN, L1CAM, NRG1, NGFR, and PDGFB. On the other hand, angiogenesis inhibitors and related genes, including IL12A, MLLT7, STAB1, and TIMP2, are downregulated. Taken together, hypoxic/normoxic preconditioning may lead to the differentiation of CD133+ cells toward endothelial lineage, which may improve the current clinical trial studies.
Collapse
Affiliation(s)
- Lee-Lee Ong
- Department of Cardiac Surgery, University of Rostock, Rostock, Germany
| | | | | | | | | | | | | | | | | | | |
Collapse
|
17
|
Aguzzi MS, Faraone D, D'Arcangelo D, De Marchis F, Toietta G, Ribatti D, Parazzoli A, Colombo P, Capogrossi MC, Facchiano A. The FGF-2-derived peptide FREG inhibits melanoma growth in vitro and in vivo. Mol Ther 2011; 19:266-273. [PMID: 20924364 PMCID: PMC3034841 DOI: 10.1038/mt.2010.211] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2010] [Accepted: 09/02/2010] [Indexed: 02/07/2023] Open
Abstract
Previous data report that fibroblast growth factor-2 (FGF-2)-derived peptide FREG potently inhibits FGF-2-dependent angiogenesis in vitro and in vivo. Here, we show that FREG inhibits up to 70% in vitro growth and invasion/migration of smooth muscle and melanoma cells. Such inhibition is mediated by platelet-derived growth factor-receptor-α (PDGF-Rα); in fact, proliferation and migration were restored upon PDGF-Rα neutralization. Further experiments demonstrated that FREG interacts with PDGF-Rα both in vitro and in vivo and stimulates its phosphorylation. We have previously shown that overexpressing PDGF-Rα strongly inhibits melanoma growth in vivo; we, therefore, hypothesized that PDGF-Rα agonists may represent a novel tool to inhibit melanoma growth in vivo. To support this hypothesis, FREG was inoculated intravenously (i.v.) in a mouse melanoma model and markedly inhibited pulmonary metastases formation. Immunohistochemical analyses showed less proliferation, less angiogenesis, and more apoptosis in metastasized lungs upon FREG treatment, as compared to untreated controls. Finally, in preliminary acute toxicity studies, FREG showed no toxicity signs in healthy animals, and neither microscopic nor macroscopic toxicity at the liver, kidney, and lungs level. Altogether, these data indicate that FREG systemic treatment strongly inhibits melanoma metastases development and indicate for the first time that agonists of PDGF-Rα may control melanoma both in vitro and in vivo.
Collapse
Affiliation(s)
- Maria S Aguzzi
- Laboratorio Patologia Vascolare, Istituto Dermopatico dell'Immacolata, IDI-IRCSS, Roma, Italy
| | | | | | | | | | | | | | | | | | | |
Collapse
|
18
|
Helmke BM, Markowski DN, Müller MH, Sommer A, Müller J, Möller C, Bullerdiek J. HMGA proteins regulate the expression of FGF2 in uterine fibroids. Mol Hum Reprod 2010; 17:135-42. [PMID: 20926602 DOI: 10.1093/molehr/gaq083] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
In human fibroids genes encoding the high-mobility proteins containing the 'AT-hook' DNA-binding motif (HMGA) are frequently affected by non-random chromosomal rearrangements. Thus, the different proteins and their derivatives resulting from these genomic rearrangements can be assumed to be involved in the genesis of these tumors by activation of largely identical downstream pathways. Constructs encoding HMGA proteins and their relevant derivatives were overexpressed in human myometrial cells, and RNA isolated from these cells was hybridized to filter arrays. Four genes were either up- or down-regulated at least 2-fold after overexpression of either of the HMGA genes and their derivatives. FGF2 (fibroblast growth factor 2) was one of these genes, and we were then able to show by microarray analyses that tumors with rearrangements of the HMGA2 locus (n = 8) expressed significantly higher levels of FGF2 than those with an apparently normal karyotype (n = 47). Accordingly, by quantitative real-time PCR uterine leiomyomas with rearrangements of the HMGA2 locus were found to express significantly higher levels of FGF2 than those with an apparently normal karyotype with a linear relationship between the expression of FGF2 and the level of HMGA2 overexpression as well as the tumor size. The results of western blot analyses confirmed these findings. Moreover, stimulation of myometrial tissue by FGF1, a strong inducer of HMGA2, leads to an increase of HMGA2 as well as FGF2 expression. In conclusion, the results contribute to the understanding of the association between the overexpression of HMGA proteins, the regulation of FGF2 expression and the size of fibroids.
Collapse
|
19
|
Leali D, Bianchi R, Bugatti A, Nicoli S, Mitola S, Ragona L, Tomaselli S, Gallo G, Catello S, Rivieccio V, Zetta L, Presta M. Fibroblast growth factor 2-antagonist activity of a long-pentraxin 3-derived anti-angiogenic pentapeptide. J Cell Mol Med 2010; 14:2109-21. [PMID: 19627396 PMCID: PMC3823002 DOI: 10.1111/j.1582-4934.2009.00855.x] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Fibroblast growth factor-2 (FGF2) plays a major role in angiogenesis. The pattern recognition receptor long-pentraxin 3 (PTX3) inhibits the angiogenic activity of FGF2. To identify novel FGF2-antagonistic peptide(s), four acetylated (Ac) synthetic peptides overlapping the FGF2-binding region PTX3-(97-110) were assessed for their FGF2-binding capacity. Among them, the shortest pentapeptide Ac-ARPCA-NH(2) (PTX3-[100-104]) inhibits the interaction of FGF2 with PTX3 immobilized to a BIAcore sensorchip and suppresses FGF2-dependent proliferation in endothelial cells, without affecting the activity of unrelated mitogens. Also, Ac-ARPCA-NH(2) inhibits angiogenesis triggered by FGF2 or by tumorigenic FGF2-overexpressing murine endothelial cells in chick and zebrafish embryos, respectively. Accordingly, the peptide hampers the binding of FGF2 to Chinese Hamster ovary cells overexpressing the tyrosine-kinase FGF receptor-1 (FGFR1) and to recombinant FGFR1 immobilized to a BIAcore sensorchip without affecting heparin interaction. In all the assays the mutated Ac-ARPSA-NH(2) peptide was ineffective. In keeping with the observation that hydrophobic interactions dominate the interface between FGF2 and the FGF-binding domain of the Ig-like loop D2 of FGFR1, amino acid substitutions in Ac-ARPCA-NH(2) and saturation transfer difference-nuclear magnetic resonance analysis of its mode of interaction with FGF2 implicate the hydrophobic methyl groups of the pentapeptide in FGF2 binding. These results will provide the basis for the design of novel PTX3-derived anti-angiogenic FGF2 antagonists.
Collapse
Affiliation(s)
- Daria Leali
- Unit of General Pathology and Immunology, Department of Biomedical Sciences and Biotechnology, School of Medicine, University of Brescia, Brescia, Italy
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
20
|
Wakamatsu A, Kimura K, Yamamoto JI, Nishikawa T, Nomura N, Sugano S, Isogai T. Identification and functional analyses of 11,769 full-length human cDNAs focused on alternative splicing. DNA Res 2009; 16:371-83. [PMID: 19880432 PMCID: PMC2780955 DOI: 10.1093/dnares/dsp022] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
We analyzed diversity of mRNA produced as a result of alternative splicing in order to evaluate gene function. First, we predicted the number of human genes transcribed into protein-coding mRNAs by using the sequence information of full-length cDNAs and 5′-ESTs and obtained 23 241 of such human genes. Next, using these genes, we analyzed the mRNA diversity and consequently sequenced and identified 11 769 human full-length cDNAs whose predicted open reading frames were different from other known full-length cDNAs. Especially, 30% of the cDNAs we identified contained variation in the transcription start site (TSS). Our analysis, which particularly focused on multiple variable first exons (FEVs) formed due to the alternative utilization of TSSs, led to the identification of 261 FEVs expressed in the tissue-specific manner. Quantification of the expression profiles of 13 genes by real-time PCR analysis further confirmed the tissue-specific expression of FEVs, e.g. OXR1 had specific TSS in brain and tumor tissues, and so on. Finally, based on the results of our mRNA diversity analysis, we have created the FLJ Human cDNA Database. From our result, it has been understood mechanisms that one gene produces suitable protein-coding transcripts responding to the situation and the environment.
Collapse
Affiliation(s)
- Ai Wakamatsu
- Graduate School of Pharmaceutical Sciences, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Kouichi Kimura
- Central Research Laboratory, Hitachi, Ltd, Kokubunji, Tokyo 185-8601, Japan
| | - Jun-ichi Yamamoto
- Reverse Proteomics Research Institute, 1-9-11 Kaji, Chiyoda-ku, Tokyo 101-0044, Japan
| | - Tetsuo Nishikawa
- Reverse Proteomics Research Institute, 1-9-11 Kaji, Chiyoda-ku, Tokyo 101-0044, Japan
| | - Nobuo Nomura
- National Institute of Advanced Industrial Science and Technology, 2-41-6 Aomi, Koto-ku, Tokyo 135-0064, Japan
| | - Sumio Sugano
- Department of Medical Genome Sciences, Graduate School of Frontier Sciences, The University of Tokyo, 4-6-1 Shiroganedai, Minato-ku, Tokyo 108-8639, Japan
| | - Takao Isogai
- Graduate School of Pharmaceutical Sciences, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
- Reverse Proteomics Research Institute, 1-9-11 Kaji, Chiyoda-ku, Tokyo 101-0044, Japan
- Corresponding author. E-mail:
| |
Collapse
|
21
|
Rusnati M, Bugatti A, Mitola S, Leali D, Bergese P, Depero LE, Presta M. Exploiting Surface Plasmon Resonance (SPR) Technology for the Identification of Fibroblast Growth Factor-2 (FGF2) Antagonists Endowed with Antiangiogenic Activity. SENSORS (BASEL, SWITZERLAND) 2009; 9:6471-503. [PMID: 22454596 PMCID: PMC3312455 DOI: 10.3390/s90806471] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/24/2009] [Revised: 08/17/2009] [Accepted: 08/19/2009] [Indexed: 12/31/2022]
Abstract
Angiogenesis, the process of new blood vessel formation, is implicated in various physiological/pathological conditions, including embryonic development, inflammation and tumor growth. Fibroblast growth factor-2 (FGF2) is a heparin-binding angiogenic growth factor involved in various physiopathological processes, including tumor neovascularization. Accordingly, FGF2 is considered a target for antiangiogenic therapies. Thus, numerous natural/synthetic compounds have been tested for their capacity to bind and sequester FGF2 in the extracellular environment preventing its interaction with cellular receptors. We have exploited surface plasmon resonance (SPR) technique in search for antiangiogenic FGF2 binders/antagonists. In this review we will summarize our experience in SPR-based angiogenesis research, with the aim to validate SPR as a first line screening for the identification of antiangiogenic compounds.
Collapse
Affiliation(s)
- Marco Rusnati
- Unit of General Pathology and Immunology, Department of Biomedical Sciences and Biotechnology, School of Medicine, University of Brescia, Brescia, 25123, Italy; E-Mails: (M.R.); (A.B.); (S.M.); (D.L.)
| | - Antonella Bugatti
- Unit of General Pathology and Immunology, Department of Biomedical Sciences and Biotechnology, School of Medicine, University of Brescia, Brescia, 25123, Italy; E-Mails: (M.R.); (A.B.); (S.M.); (D.L.)
| | - Stefania Mitola
- Unit of General Pathology and Immunology, Department of Biomedical Sciences and Biotechnology, School of Medicine, University of Brescia, Brescia, 25123, Italy; E-Mails: (M.R.); (A.B.); (S.M.); (D.L.)
| | - Daria Leali
- Unit of General Pathology and Immunology, Department of Biomedical Sciences and Biotechnology, School of Medicine, University of Brescia, Brescia, 25123, Italy; E-Mails: (M.R.); (A.B.); (S.M.); (D.L.)
| | - Paolo Bergese
- Chemistry for Technologies Laboratory and Department of Mechanical and Industrial Engineering, School of Engineering, University of Brescia, Brescia, 25123, Italy; E-Mails: (P.B.); (L.E.D.)
| | - Laura E. Depero
- Chemistry for Technologies Laboratory and Department of Mechanical and Industrial Engineering, School of Engineering, University of Brescia, Brescia, 25123, Italy; E-Mails: (P.B.); (L.E.D.)
| | - Marco Presta
- Unit of General Pathology and Immunology, Department of Biomedical Sciences and Biotechnology, School of Medicine, University of Brescia, Brescia, 25123, Italy; E-Mails: (M.R.); (A.B.); (S.M.); (D.L.)
| |
Collapse
|
22
|
Totta P, De Cristofaro R, Giampietri C, Aguzzi MS, Faraone D, Capogrossi MC, Facchiano A. Thrombin-mediated impairment of fibroblast growth factor-2 activity. FEBS J 2009; 276:3277-89. [PMID: 19438723 DOI: 10.1111/j.1742-4658.2009.07042.x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Thrombin generation increases in several pathological conditions, including cancer, thromboembolism, diabetes and myeloproliferative syndromes. During tumor development, thrombin levels increase along with several other molecules, including cytokines and angiogenic factors. Under such conditions, it is reasonable to predict that thrombin may recognize new low-affinity substrates that usually are not recognized under low-expression levels conditions. In the present study, we hypothesized that fibroblast growth factor (FGF)-2 may be cleaved by thrombin and that such action may lead to an impairment of its biological activity. The evidence collected in the present study indicates that FGF-2-induced proliferation and chemotaxis/invasion of SK-MEL-110 human melanoma cells were significantly reduced when FGF-2 was pre-incubated with active thrombin. The inhibition of proliferation was not influenced by heparin. Phe-Pro-Arg-chloromethyl ketone, a specific inhibitor of the enzymatic activity of thrombin, abolished the thrombin-induced observed effects. Accordingly, both FGF-2-binding to cell membranes as well as FGF-2-induced extracellular signal-regulated kinase phosphorylation were decreased in the presence of thrombin. Finally, HPLC analyses demonstrated that FGF-2 is cleaved by thrombin at the peptide bond between residues Arg42 and Ile43 of the mature human FGF-2 sequence. The apparent k(cat)/K(m) of FGF-2 hydrolysis was 1.1 x 10(4) M(-1) x s(-1), which is comparable to other known low-affinity thrombin substrates. Taken together, these results demonstrate that thrombin digests FGF-2 at the site Arg42-Ile43 and impairs FGF-2 activity in vitro, indicating that FGF-2 is a novel thrombin substrate.
Collapse
Affiliation(s)
- Pierangela Totta
- Laboratorio di Patologia Vascolare, IDI-IRCCS, Istituto Dermopatico dell'Immacolata-Istituto di Ricovero e Cura a Carattere Scientifico, Rome, Italy
| | | | | | | | | | | | | |
Collapse
|
23
|
Nickel W, Seedorf M. Unconventional Mechanisms of Protein Transport to the Cell Surface of Eukaryotic Cells. Annu Rev Cell Dev Biol 2008; 24:287-308. [PMID: 18590485 DOI: 10.1146/annurev.cellbio.24.110707.175320] [Citation(s) in RCA: 206] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
- Walter Nickel
- Heidelberg University Biochemistry Center (BZH) 69120 Heidelberg, Germany
| | - Matthias Seedorf
- Zentrum für Molekulare Biologie der Universität Heidelberg (ZMBH), 69120 Heidelberg, Germany;
| |
Collapse
|
24
|
Rose K, Pallast S, Klumpp S, Krieglstein J. ATP-binding on fibroblast growth factor 2 partially overlaps with the heparin-binding domain. J Biochem 2008; 144:343-7. [PMID: 18511454 DOI: 10.1093/jb/mvn072] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Fibroblast growth factor 2 (FGF2), an intensively studied heparin-binding cytokine, is an important modulator of cell growth and differentiation under both physiological and pathophysiological conditions. It has been shown recently that ATP binds to FGF2 and that this binding is crucial for its biological function. In this study we demonstrated that divalent cations were not necessary for binding of ATP to FGF2, but it could be demonstrated that heparin blocked the labelling of FGF2 with ATP indicating an involvement of the heparin-binding domain (aa 128-144) in ATP-binding. FGF2, bound to Heparin Sepharose, could be eluted with ATP and GTP, but not with cAMP, AMP or ADP. Successive mutation of positively charged amino acid residues located in the heparin-binding domain drastically reduced the signal intensity of [gamma-(32)P]ATP labelled FGF2 indicating that this domain is not only important for heparin binding to FGF2 but also for ATP-binding.
Collapse
Affiliation(s)
- Karsten Rose
- Institut für Pharmazeutische und Medizinische Chemie, Westfälische Wilhelms-Universität Münster, Hittorfstr. 58-62, 48149 Münster, Germany
| | | | | | | |
Collapse
|
25
|
Rose K, Kriha D, Pallast S, Junker V, Klumpp S, Krieglstein J. Basic fibroblast growth factor: lysine 134 is essential for its neuroprotective activity. Neurochem Int 2007; 51:25-31. [PMID: 17524524 DOI: 10.1016/j.neuint.2007.03.011] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2007] [Revised: 03/26/2007] [Accepted: 03/28/2007] [Indexed: 01/24/2023]
Abstract
Basic fibroblast growth factor (bFGF) is a heparin-binding growth factor known to cause cell proliferation, angiogenesis and neuroprotection. We have performed site-directed mutagenesis to identify the amino acids that are essential for heparin/growth factor interaction and for neuroprotection. Binding to heparin-acrylic beads was markedly reduced when lysine in position 134 of bFGF was replaced by alanine. Wildtype (wt)-bFGF was shown to protect rat primary cultures of embryonic hippocampal neurons against damage caused by staurosporine and to reduce the infarct size in mice after focal cerebral ischemia. These neuroprotective effects of wt-bFGF could not be shown for the mutant bFGF(K134A). Furthermore, phosphorylation of Akt and ERK1/2 was significantly reduced in cultured neurons treated with bFGF(K134A) indicating diminished intracellular signaling compared to neurons treated with wt-bFGF. In conclusion, lysine at position 134 of bFGF is essential for bFGF to bind heparin, then to interact with its receptor and, subsequently, to protect neurons against damage.
Collapse
Affiliation(s)
- Karsten Rose
- Institut für Pharmazeutische und Medizinische Chemie, Westfälische Wilhelms-Universität, Hittorfstr. 58-62, D-48149 Münster, Germany
| | | | | | | | | | | |
Collapse
|
26
|
Faraone D, Aguzzi MS, Ragone G, Russo K, Capogrossi MC, Facchiano A. Heterodimerization of FGF-receptor 1 and PDGF-receptor-alpha: a novel mechanism underlying the inhibitory effect of PDGF-BB on FGF-2 in human cells. Blood 2005; 107:1896-902. [PMID: 16322476 DOI: 10.1182/blood-2005-04-1524] [Citation(s) in RCA: 38] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Previous evidence has shown that platelet-derived growth factor-BB (PDGF-BB) and fibroblast growth factor-2 (FGF-2) directly interact with high affinity, leading to potent reciprocal inhibitory effects on bovine endothelial cells and rat vascular smooth muscle cells. In this study, we report that PDGF-BB inhibits a series of FGF-2-induced events, such as proliferation of human umbilical vein endothelial cells (HUVECs), FGF-2 cellular internalization, phosphorylation of intracellular signaling factors including p38, rac1/cdc42, MKK4, and MKK3/6, and phosphorylation of FGF-receptor 1 (FGF-R1). PDGF-receptor-alpha (PDGF-Ralpha) was found to mediate PDGF-BB inhibitory effects because its neutralization fully restored FGF-2 mitogenic activity and internalization. Additional biochemical analyses, coimmunoprecipitation experiments, and FRET analysis showed that FGF-R1 and PDGF-Ralpha directly interact in vitro and in vivo and that this interaction is somehow increased in the presence of the corresponding ligands FGF-2 and PDGF-BB. These results suggest that FGF-R1/PDGF-Ralpha heterodimerization may represent a novel endogenous mechanism to modulate the action of these receptors and their ligands and to control endothelial cell function.
Collapse
Affiliation(s)
- Debora Faraone
- Laboratorio di Patologia Vascolare, Istituto Dermopatico della Immacolata, IDI-IRCCS, Via Monti di Creta 104, 00167 Rome, Italy
| | | | | | | | | | | |
Collapse
|
27
|
Klumpp S, Kriha D, Bechmann G, Maassen A, Maier S, Pallast S, Hoell P, Krieglstein J. Phosphorylation of the growth factors bFGF, NGF and BDNF: a prerequisite for their biological activity. Neurochem Int 2005; 48:131-7. [PMID: 16242215 DOI: 10.1016/j.neuint.2005.08.009] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2005] [Accepted: 08/25/2005] [Indexed: 01/19/2023]
Abstract
The aim of this work was to test whether growth factors such as basic fibroblast growth factor (bFGF), nerve growth factor (NGF) and brain-derived neurotrophic factor (BDNF) undergo autophosphorylation and whether this affects their biological activity. Incubation of those growth factors with [gamma-(32)P]ATP resulted in phosphorylation in vitro. The phosphate bond was resistant to alkaline pH, yet acid-labile. Addition of alkaline phosphatase resulted in time and protein dependent dephosphorylation. Concomitantly, alkaline phosphatase abolished the neuroprotective effect of those growth factors upon oxygen and glucose deprivation and upon staurosporine-induced cell death. For those studies, we were using primary cultures of cortical and hippocampal neurons from embryonic and neonatal rats. Incubation of bFGF with non-hydrolyzable ATP-gammaS resulted in phosphorylation and in neuroprotection resistant to alkaline phosphatase. We conclude that bFGF, NGF and BDNF undergo autophosphorylation on site(s) other than serine, threonine, tyrosine and/or ATP-binding, and that this binding of phosphate is essential for neuroprotection in vivo.
Collapse
Affiliation(s)
- Susanne Klumpp
- Institut für Pharmazeutische & Medizinische Chemie, Westfälische Wilhelms-Universität, Hittorfstr. 58-62, D-48149 Münster, Germany.
| | | | | | | | | | | | | | | |
Collapse
|
28
|
Soulet F, Bailly K, Roga S, Lavigne AC, Amalric F, Bouche G. Exogenously Added Fibroblast Growth Factor 2 (FGF-2) to NIH3T3 CellsInteracts with Nuclear Ribosomal S6 Kinase 2 (RSK2) in a Cell Cycle-dependentManner. J Biol Chem 2005; 280:25604-10. [PMID: 15879597 DOI: 10.1074/jbc.m500232200] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Fibroblast growth factor 2 (FGF-2) has been detected in the nuclei of many tissues and cell lines. Here we demonstrate that FGF-2 added exogenously to NIH3T3 cells enters the nucleus and interacts with the nuclear active 90-kDa ribosomal S6 kinase 2 (RSK2) in a cell cycle-dependent manner. By using purified proteins, FGF-2 is shown to directly interact through two separate domains with two RSK2 domains on both sides of the hydrophobic motif, namely the NH2-terminal kinase domain (residues 360-381) by amino acid Ser-117 and the COOH-terminal kinase domain (residues 388-400) by amino acids Leu-127 and Lys-128. Moreover, this interaction leads to maintenance of the sustained activation of RSK2 in G1 phase of the cell cycle. FGF-2 mutants (FGF-2 S117A, FGF-2 L127A, and FGF-2 K128A) that fail to interact in vitro with RSK2 fail to maintain a sustained RSK2 activity in vivo.
Collapse
Affiliation(s)
- Fabienne Soulet
- Laboratoire de Biologie Vasculaire, Institut de Pharmacologie et de Biologie Structurale, Unité Mixte de Recherche 5089, 205 Route de Narbonne, 31077 Toulouse, France
| | | | | | | | | | | |
Collapse
|
29
|
Abstract
In the year 2003 there was a 17% increase in the number of publications citing work performed using optical biosensor technology compared with the previous year. We collated the 962 total papers for 2003, identified the geographical regions where the work was performed, highlighted the instrument types on which it was carried out, and segregated the papers by biological system. In this overview, we spotlight 13 papers that should be on everyone's 'must read' list for 2003 and provide examples of how to identify and interpret high-quality biosensor data. Although we still find that the literature is replete with poorly performed experiments, over-interpreted results and a general lack of understanding of data analysis, we are optimistic that these shortcomings will be addressed as biosensor technology continues to mature.
Collapse
Affiliation(s)
- Rebecca L Rich
- Center for Biomolecular Interaction Analysis, University of Utah, Salt Lake City, UT 84132, USA
| | | |
Collapse
|
30
|
Hamada Y, Nokihara K, Okazaki M, Fujitani W, Matsumoto T, Matsuo M, Umakoshi Y, Takahashi J, Matsuura N. Angiogenic activity of osteopontin-derived peptide SVVYGLR. Biochem Biophys Res Commun 2003; 310:153-7. [PMID: 14511663 DOI: 10.1016/j.bbrc.2003.09.001] [Citation(s) in RCA: 70] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Angiogenesis plays an important role in various pathological conditions as well as some physiological processes. Although a number of soluble angiogenic factors have been reported, extracellular matrix also has crucial effect on angiogenesis through interaction with endothelial cells. Since recent reports showed osteopontin had some angiogenic activity, the effect of the SVVYGLR peptide, novel binding motif in osteopontin molecule, on angiogenesis was examined in this study. Synthetic peptide SVVYGLR did not have proliferative effect on endothelial cells but adhesion and migration activity to endothelial cells. Furthermore, SVVYGLR had as potent activity for tube formation in three-dimensional collagen gel as vascular endothelial growth factor which is known to be the strongest angiogenic factor. Electron microscopical analysis showed a number of microvilli on the endothelial luminar surface and tight junction formation in the luminar intercellular border between endothelial cells, indicating SVVYGLR induced cell porarity and differentiation of endothelial cells. This small peptide might be expected to stimulate angiogenesis to improve some ischemic conditions in the future because of some advantages due to smaller molecular weight.
Collapse
Affiliation(s)
- Yoshinosuke Hamada
- Department of Pathology, School of Allied Health Sciences, Faculty of Medicine, Osaka University, Osaka 565-0871, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|