1
|
Liu Y, Wei M, Li G, Zhao Y, Yan X, Wang S, Song X, Wang Z, Huang L. Isolation, structural characterization of natural chondroitin sulfate oligosaccharides and their binding study with anti-angiogenic factors. Carbohydr Polym 2025; 353:123262. [PMID: 39914977 DOI: 10.1016/j.carbpol.2025.123262] [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: 09/26/2024] [Revised: 01/06/2025] [Accepted: 01/10/2025] [Indexed: 05/07/2025]
Abstract
Drugs that inhibit tumor angiogenesis, promote vascular normalization and improve the tumor microenvironment. However, their application is limited by adaptive or compensatory resistance. Chondroitin sulfate (CS) regulates numerous proteins including pro-angiogenic growth factors, for whom binding affinity depends on sulfation of CS. In this study, we aimed to determine how sulfation of natural tetrasaccharides and hexasaccharides of CS affected binding to the vascular endothelial growth factor (VEGF-A) and fibroblast growth factor 2 (FGF-2). Twenty-eight CS oligosaccharide isomers were obtained by preparative HPLC, tagged with the AEAB fluorescent linker, and identified using an improved chemical derivatization strategy combined with tandem mass spectrometry. CS oligosaccharide microarrays revealed that VEGF-A and FGF-2 bound preferentially to highly sulfated CS, and the GalNAc(4S)GlcA(2S)GalNAc(6S) sequence was found to be indispensable for binding to these proteins. By integrating glycan microarrays with computational modeling, this study revealed the relationship between the structure of CS and its interactions with pro-angiogenic factors. The degree and the specific sulfation patterns on CS should be taken into account when designing anti-angiogenic drugs.
Collapse
Affiliation(s)
- Yuxia Liu
- Shaanxi Natural Carbohydrate Resource Engineering Research Center, College of Food Science and Technology, Northwest University, Xi'an 710069, PR China
| | - Ming Wei
- Shaanxi Natural Carbohydrate Resource Engineering Research Center, College of Food Science and Technology, Northwest University, Xi'an 710069, PR China; School of Marine and Bioengineering, YanCheng Institute of Technology, Yancheng 224051, PR China
| | - Guo Li
- Shaanxi Natural Carbohydrate Resource Engineering Research Center, College of Food Science and Technology, Northwest University, Xi'an 710069, PR China
| | - Yilong Zhao
- Shaanxi Natural Carbohydrate Resource Engineering Research Center, College of Food Science and Technology, Northwest University, Xi'an 710069, PR China
| | - Xiuzhen Yan
- Shaanxi Natural Carbohydrate Resource Engineering Research Center, College of Food Science and Technology, Northwest University, Xi'an 710069, PR China
| | - Shukai Wang
- Shaanxi Natural Carbohydrate Resource Engineering Research Center, College of Food Science and Technology, Northwest University, Xi'an 710069, PR China
| | - Xuezheng Song
- Department of Biochemistry, Emory University School of Medicine, Atlanta, GA 30322, United States of America
| | - Zhongfu Wang
- Shaanxi Natural Carbohydrate Resource Engineering Research Center, College of Food Science and Technology, Northwest University, Xi'an 710069, PR China.
| | - Linjuan Huang
- Shaanxi Natural Carbohydrate Resource Engineering Research Center, College of Food Science and Technology, Northwest University, Xi'an 710069, PR China.
| |
Collapse
|
2
|
Hiepen C, Benamar M, Barrasa-Fano J, Condor M, Ilhan M, Münch J, Hastar N, Kerkhoff Y, Harms GS, Mielke T, Koenig B, Block S, Rocks O, Abdelilah-Seyfried S, Van Oosterwyck H, Knaus P. Endothelial tip-cell position, filopodia formation and biomechanics require BMPR2 expression and signaling. Commun Biol 2025; 8:21. [PMID: 39779836 PMCID: PMC11711618 DOI: 10.1038/s42003-024-07431-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2024] [Accepted: 12/19/2024] [Indexed: 01/11/2025] Open
Abstract
Blood vessel formation relies on biochemical and mechanical signals, particularly during sprouting angiogenesis when endothelial tip cells (TCs) guide sprouting through filopodia formation. The contribution of BMP receptors in defining tip-cell characteristics is poorly understood. Our study combines genetic, biochemical, and molecular methods together with 3D traction force microscopy, which reveals an essential role of BMPR2 for actin-driven filopodia formation and mechanical properties of endothelial cells (ECs). Targeting of Bmpr2 reduced sprouting angiogenesis in zebrafish and BMPR2-deficient human ECs formed fewer filopodia, affecting cell migration and actomyosin localization. Spheroid assays revealed a reduced sprouting of BMPR2-deficient ECs in fibrin gels. Even more strikingly, in mosaic spheroids, BMPR2-deficient ECs failed to acquire tip-cell positions. Yet, 3D traction force microscopy revealed that these distinct cell behaviors of BMPR2-deficient tip cells cannot be explained by differences in force-induced matrix deformations, even though these cells adopted distinct cone-shaped morphologies. Notably, BMPR2 positively regulates local CDC42 activity at the plasma membrane to promote filopodia formation. Our findings reveal that BMPR2 functions as a nexus integrating biochemical and biomechanical processes crucial for TCs during angiogenesis.
Collapse
Affiliation(s)
- Christian Hiepen
- Freie Universität Berlin, Institute for Chemistry and Biochemistry, Thielallee 63, 14195, Berlin, Germany.
- Westphalian University of Applied Sciences, August-Schmidt-Ring 10, 45665, Recklinghausen, Germany.
| | - Mounir Benamar
- Freie Universität Berlin, Institute for Chemistry and Biochemistry, Thielallee 63, 14195, Berlin, Germany
| | - Jorge Barrasa-Fano
- KU Leuven, Department of Mechanical Engineering, Biomechanics section, Leuven, Celestijnenlaan 300 C, 3001, Leuven, Belgium
| | - Mar Condor
- KU Leuven, Department of Mechanical Engineering, Biomechanics section, Leuven, Celestijnenlaan 300 C, 3001, Leuven, Belgium
| | - Mustafa Ilhan
- Freie Universität Berlin, Institute for Chemistry and Biochemistry, Thielallee 63, 14195, Berlin, Germany
- Berlin School of Integrative Oncology, Augustenburger Platz 1, D-13353, Berlin, Germany
| | - Juliane Münch
- Universität Potsdam, Institute of Biochemistry and Biology, Karl-Liebknecht Strasse 24-25, 14476, Potsdam-Golm, Germany
| | - Nurcan Hastar
- Freie Universität Berlin, Institute for Chemistry and Biochemistry, Thielallee 63, 14195, Berlin, Germany
| | - Yannic Kerkhoff
- Freie Universität Berlin, Institute for Chemistry and Biochemistry, Thielallee 63, 14195, Berlin, Germany
| | - Gregory S Harms
- Universitätsmedizin, Johannes Gutenberg-Universität Mainz, Cell Biology Unit, Imaging Core Facility and the Research Center for Immune Intervention, Langenbeckstraße 1, 55131, Mainz, Germany
| | - Thorsten Mielke
- Max-Planck-Institute for Molecular Genetics, Microscopy & Cryo-Electron Microscopy, Ihnestraße 63-73, 14195, Berlin, Germany
| | - Benjamin Koenig
- Freie Universität Berlin, Institute for Chemistry and Biochemistry, Thielallee 63, 14195, Berlin, Germany
- Leibniz Forschungsinstitut für Molekulare Pharmakologie (FMP), Berlin, Germany
| | - Stephan Block
- Freie Universität Berlin, Institute for Chemistry and Biochemistry, Thielallee 63, 14195, Berlin, Germany
| | - Oliver Rocks
- Charité - Universitätsmedizin Berlin, Systemic Cell Dynamics, Charitéplatz 1, 10117, Berlin, Germany
| | - Salim Abdelilah-Seyfried
- Universität Potsdam, Institute of Biochemistry and Biology, Karl-Liebknecht Strasse 24-25, 14476, Potsdam-Golm, Germany
| | - Hans Van Oosterwyck
- KU Leuven, Department of Mechanical Engineering, Biomechanics section, Leuven, Celestijnenlaan 300 C, 3001, Leuven, Belgium
- KU Leuven, Prometheus Division of Skeletal Tissue Engineering, Leuven, Belgium
| | - Petra Knaus
- Freie Universität Berlin, Institute for Chemistry and Biochemistry, Thielallee 63, 14195, Berlin, Germany.
| |
Collapse
|
3
|
Yuan Y, Dai Y, Wang J, Shen G, Gai Y, Dong Q, Liu L, Zhu X, Jiang D, Xi L, Dai J, Li F. Identification of a Novel Vascular Endothelial Growth Factor Receptor-3-Targeting Peptide for Molecular Imaging of Metastatic Lymph Nodes. Bioconjug Chem 2024; 35:1843-1858. [PMID: 39469784 DOI: 10.1021/acs.bioconjchem.4c00464] [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: 10/30/2024]
Abstract
Because of the insidious nature of lymphatic metastatic cancer, accurate imaging tracing is very difficult to achieve in the clinic. Previous studies have developed the LARGR peptide (named TMVP1) as a radiotracer for vascular endothelial growth factor receptor-3 (VEGFR-3) imaging in cancer. However, its affinity for the target remains insufficient, resulting in low imaging sensitivity. In this study, we identified a high-affinity VEGFR-3 targeting peptide, named TMVP1446, using a multiplex screening platform. TMVP1446 demonstrated a dissociation constant of 8.97 × 10-8 M. Both in vitro and in vivo assays confirmed that fluorescently labeled TMVP1446 specifically bound to VEGFR-3. In a 4T1-luciferase tumor mouse model, cyanine 7-labeled TMVP1446 effectively discriminated between contralateral normal lymph nodes (c-LN) and cancer-metastatic sentinel lymph nodes (m-SLN). To evaluate the potential of TMVP1446, we developed a novel VEGFR-3 positron emission tomography radiotracer ([68Ga]Ga-DOTA-TMVP1446) for cancer-m-SLN imaging. [68Ga]Ga-DOTA-TMVP1446 accurately detected and assessed the status of lymph node metastasis, even in micrometastatic tumors, in the B16-F10 mouse tumor model. These findings suggest that TMVP1446 has great potential for advancing VEGFR-3 molecular imaging and metastatic sentinel lymph node imaging.
Collapse
Affiliation(s)
- Yuan Yuan
- Department of Obstetrics and Gynecology, National Clinical Research Center for Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
- Key Laboratory of Cancer Invasion and Metastasis (Ministry of Education), Hubei Key Laboratory of Tumor Invasion and Metastasis, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Yilin Dai
- Department of Obstetrics and Gynecology, National Clinical Research Center for Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
- Key Laboratory of Cancer Invasion and Metastasis (Ministry of Education), Hubei Key Laboratory of Tumor Invasion and Metastasis, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Jing Wang
- Department of Obstetrics and Gynecology, National Clinical Research Center for Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
- Key Laboratory of Cancer Invasion and Metastasis (Ministry of Education), Hubei Key Laboratory of Tumor Invasion and Metastasis, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Guangyang Shen
- Department of Obstetrics and Gynecology, National Clinical Research Center for Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
- Key Laboratory of Cancer Invasion and Metastasis (Ministry of Education), Hubei Key Laboratory of Tumor Invasion and Metastasis, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Yongkang Gai
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Qingjian Dong
- Department of Nuclear Medicine and PET, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430034, China
| | - Luoxia Liu
- Department of Nuclear Medicine and PET, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430034, China
| | - Xiaohua Zhu
- Department of Nuclear Medicine and PET, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430034, China
| | - Dawei Jiang
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Ling Xi
- Department of Obstetrics and Gynecology, National Clinical Research Center for Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
- Key Laboratory of Cancer Invasion and Metastasis (Ministry of Education), Hubei Key Laboratory of Tumor Invasion and Metastasis, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Jun Dai
- Department of Obstetrics and Gynecology, National Clinical Research Center for Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
- Key Laboratory of Cancer Invasion and Metastasis (Ministry of Education), Hubei Key Laboratory of Tumor Invasion and Metastasis, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Fei Li
- Department of Obstetrics and Gynecology, National Clinical Research Center for Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
- Key Laboratory of Cancer Invasion and Metastasis (Ministry of Education), Hubei Key Laboratory of Tumor Invasion and Metastasis, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| |
Collapse
|
4
|
Sánchez-Martínez C, Grueso E, Calvo-López T, Martinez-Ortega J, Ruiz A, Almendral JM. VEGF-Virus Interactions: Pathogenic Mechanisms and Therapeutic Applications. Cells 2024; 13:1815. [PMID: 39513922 PMCID: PMC11545703 DOI: 10.3390/cells13211815] [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: 09/03/2024] [Revised: 10/16/2024] [Accepted: 10/22/2024] [Indexed: 11/16/2024] Open
Abstract
Many types of viruses directly or indirectly target the vascular endothelial growth factor (VEGF) system, which is a central regulator of vasculogenesis and angiogenesis in physiological homeostasis, causing diverse pathologies. Other viruses have been developed into effective therapeutic tools for VEGF modulation in conditions such as cancer and eye diseases. Some viruses may alter the levels of VEGF in the pathogenesis of respiratory syndromes, or they may encode VEGF-like factors, promoting vascular disruption and angiogenesis to enable viruses' systemic spread. Oncogenic viruses may express interactive factors that perturb VEGF's functional levels or downstream signaling, which increases the neovascularization and metastasis of tumors. Furthermore, many viruses are being developed as therapeutic vectors for vascular pathologies in clinical trials. Major examples are those viral vectors that inhibit the role of VEGF in the neovascularization required for cancer progression; this is achieved through the induction of immune responses, by exposing specific peptides that block signaling or by expressing anti-VEGF and anti-VEGF receptor-neutralizing antibodies. Other viruses have been engineered into effective pro- or anti-angiogenesis multitarget vectors for neovascular eye diseases, paving the way for therapies with improved safety and minimal side effects. This article critically reviews the large body of literature on these issues, highlighting those contributions that describe the molecular mechanisms, thus expanding our understanding of the VEGF-virus interactions in disease and therapy. This could facilitate the clinical use of therapeutic virus vectors in precision medicine for the VEGF system.
Collapse
Affiliation(s)
- Cristina Sánchez-Martínez
- Biosciences Research Institute, School of Experimental Sciences, Universidad Francisco de Vitoria, Pozuelo de Alarcón, 28223 Madrid, Spain; (C.S.-M.); (E.G.)
| | - Esther Grueso
- Biosciences Research Institute, School of Experimental Sciences, Universidad Francisco de Vitoria, Pozuelo de Alarcón, 28223 Madrid, Spain; (C.S.-M.); (E.G.)
| | - Tania Calvo-López
- Centro de Biología Molecular Severo Ochoa (CSIC-UAM), Cantoblanco, 28049 Madrid, Spain or (T.C.-L.); (J.M.-O.); (A.R.)
- Departamento de Biología Molecular, Universidad Autónoma de Madrid, Cantoblanco, 28049 Madrid, Spain
- Department of Biomedicine, Centro de Investigaciones Biológicas Margarita Salas (CSIC), Ramiro de Maeztu 9, 28040 Madrid, Spain
| | - Jorge Martinez-Ortega
- Centro de Biología Molecular Severo Ochoa (CSIC-UAM), Cantoblanco, 28049 Madrid, Spain or (T.C.-L.); (J.M.-O.); (A.R.)
- Departamento de Biología Molecular, Universidad Autónoma de Madrid, Cantoblanco, 28049 Madrid, Spain
| | - Ana Ruiz
- Centro de Biología Molecular Severo Ochoa (CSIC-UAM), Cantoblanco, 28049 Madrid, Spain or (T.C.-L.); (J.M.-O.); (A.R.)
- Departamento de Biología Molecular, Universidad Autónoma de Madrid, Cantoblanco, 28049 Madrid, Spain
| | - José M. Almendral
- Centro de Biología Molecular Severo Ochoa (CSIC-UAM), Cantoblanco, 28049 Madrid, Spain or (T.C.-L.); (J.M.-O.); (A.R.)
- Departamento de Biología Molecular, Universidad Autónoma de Madrid, Cantoblanco, 28049 Madrid, Spain
| |
Collapse
|
5
|
Durak S, Sutova HE, Ceylan R, Aciksari A, Yetisgin AA, Onder Tokuc E, Kutlu O, Karabas VL, Cetinel S. A Nanogel Formulation of Anti-VEGF Peptide for Ocular Neovascularization Treatment. ACS APPLIED BIO MATERIALS 2024; 7:6001-6013. [PMID: 39167547 DOI: 10.1021/acsabm.4c00585] [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] [Indexed: 08/23/2024]
Abstract
Age-related macular degeneration (AMD) is an eye disorder that can lead to visual impairment in elder patients, and current treatments include repeated injections of monoclonal antibody-based antivascular endothelial growth factor (anti-VEGF) agents. This study investigates the potential of a nanoformulation of a peptide anti-VEGF molecule for neovascular AMD. Anti-VEGF peptide HRHTKQRHTALH (HRH), which has high affinity to VEGF-Fc receptor, was used as the bioactive agent to control neovascularization of the retina. The nanoformulation consisting of hyaluronic acid nanogel was generated by incorporating divinyl sulfone and cholesterol to increase the stability and control the size of the nanodrug. The encapsulation efficacy of nanogel was 65%, and drug release was 34.72% at the end of 192 h. Obtained nanogels were efficiently internalized in 15 min by human umbilical vascular endothelial cells (HUVECs) and ARPE-19 cells, and results indicate that nanoformulation is not toxic to ARPE-19 cells, whereas it inhibits HUVEC proliferation owing to anti-VEGF peptide in the nanogel structure. In the coculture experiment in which retinal penetration was modeled, it was observed that the nanogel reached HUVECs and negatively affected their proliferation without disturbing the monolayer of ARPE-19 cells. In vivo experiments with chick chorioallantoic membrane revealed that nanogel formulation has higher antiangiogenesis activity compared to free HRH. Additionally, in an oxygen-induced retinopathy model, the excessive growth of blood vessels was notably suppressed in mice treated with HRH-loaded nanogel. This research indicates that nanogels formulated in this study are promising candidates as a topical treatment for AMD.
Collapse
Affiliation(s)
- Saliha Durak
- Nanotechnology Research and Application Center (SUNUM), Sabanci University, Istanbul 34956, Turkey
- Faculty of Engineering and Natural Sciences, Molecular Biology, Genetics and Bioengineering Program, Sabanci University, Istanbul 34956, Turkey
| | - Hande Eda Sutova
- Nanotechnology Research and Application Center (SUNUM), Sabanci University, Istanbul 34956, Turkey
- Faculty of Engineering and Natural Sciences, Molecular Biology, Genetics and Bioengineering Program, Sabanci University, Istanbul 34956, Turkey
| | - Ramazan Ceylan
- Nanotechnology Research and Application Center (SUNUM), Sabanci University, Istanbul 34956, Turkey
- Faculty of Engineering and Natural Sciences, Sabanci University, Istanbul 34956, Turkey
| | - Aysegul Aciksari
- Nanotechnology Research and Application Center (SUNUM), Sabanci University, Istanbul 34956, Turkey
| | - Abuzer Alp Yetisgin
- Nanotechnology Research and Application Center (SUNUM), Sabanci University, Istanbul 34956, Turkey
- Faculty of Engineering and Natural Sciences, Materials Science and Nano-Engineering Program, Sabanci University, Istanbul 34956, Turkey
| | - Ecem Onder Tokuc
- Department of Ophthalmology, Kocaeli University School of Medicine, Kocaeli 41001, Turkey
| | - Ozlem Kutlu
- Nanotechnology Research and Application Center (SUNUM), Sabanci University, Istanbul 34956, Turkey
- Faculty of Engineering and Natural Sciences, Molecular Biology, Genetics and Bioengineering Program, Sabanci University, Istanbul 34956, Turkey
| | - Veysel Levent Karabas
- Department of Ophthalmology, Kocaeli University School of Medicine, Kocaeli 41001, Turkey
| | - Sibel Cetinel
- Nanotechnology Research and Application Center (SUNUM), Sabanci University, Istanbul 34956, Turkey
- Faculty of Engineering and Natural Sciences, Molecular Biology, Genetics and Bioengineering Program, Sabanci University, Istanbul 34956, Turkey
| |
Collapse
|
6
|
Pu G, Liang Z, Shi J, Tao Y, Lu P, Qing H, Zhang J. Enhancing the Inhibition of Corneal Neovascularization Efficacy by Self-Assembled into Supramolecular Hydrogel of Anti-Angiogenic Peptide. Int J Nanomedicine 2024; 19:7605-7616. [PMID: 39081898 PMCID: PMC11287374 DOI: 10.2147/ijn.s465965] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Accepted: 07/10/2024] [Indexed: 08/02/2024] Open
Abstract
Background Corneal neovascularization (CNV) is a common eye disease that leads to blindness. New treatment strategies are urgently needed due to the limitations of current treatment methods. Methods We report the synthesis of peptide Nap-FFEEPCAIWF ( Comp.3 ) via chemical conjugation of Nap-FFEE ( Comp.2 ) to antiangiogenic peptide PCAIWF (Comp.1). Comp.3 self-assembled into a hydrogel ( gel of 3 ) composed of nanofibers, which enhanced the antiangiogenic function of the epitope. Results We developed a novel peptide with an amphiphilic framework, Comp.3 , which could self-assemble into a supramolecular hydrogel with a well-ordered nanofiber structure. The nanofibers exhibited good biocompatibility with corneal epithelial cells, presenting a promising strategy to enhance the efficacy of free peptide-based drugs in the treatment of ocular vascular diseases, such as CNV and other angiogenesis-related diseases. Conclusion Nap-FFEEPCAIWF nanofibers provide an alternative approach to enhancing the therapeutic efficiency of free peptide-based drugs against ocular vascular diseases.
Collapse
Affiliation(s)
- Guojuan Pu
- Henan Eye Hospital, Henan Eye Institute, Henan Provincial People’s Hospital, Zhengzhou, 450003, People’s Republic of China
- People’s Hospital of Zhengzhou University, Zhengzhou, 450003, People’s Republic of China
| | - Zhen Liang
- Henan Eye Hospital, Henan Eye Institute, Henan Provincial People’s Hospital, Zhengzhou, 450003, People’s Republic of China
- People’s Hospital of Zhengzhou University, Zhengzhou, 450003, People’s Republic of China
| | - Jieran Shi
- People’s Hospital of Zhengzhou University, Zhengzhou, 450003, People’s Republic of China
| | - Yuan Tao
- Henan Eye Hospital, Henan Eye Institute, Henan Provincial People’s Hospital, Zhengzhou, 450003, People’s Republic of China
- People’s Hospital of Zhengzhou University, Zhengzhou, 450003, People’s Republic of China
| | - Ping Lu
- Henan Eye Hospital, Henan Eye Institute, Henan Provincial People’s Hospital, Zhengzhou, 450003, People’s Republic of China
- People’s Hospital of Zhengzhou University, Zhengzhou, 450003, People’s Republic of China
| | - Huiling Qing
- Henan Eye Hospital, Henan Eye Institute, Henan Provincial People’s Hospital, Zhengzhou, 450003, People’s Republic of China
- People’s Hospital of Zhengzhou University, Zhengzhou, 450003, People’s Republic of China
| | - Junjie Zhang
- Henan Eye Hospital, Henan Eye Institute, Henan Provincial People’s Hospital, Zhengzhou, 450003, People’s Republic of China
- People’s Hospital of Zhengzhou University, Zhengzhou, 450003, People’s Republic of China
| |
Collapse
|
7
|
Giordano RJ, Alecrim LC. Protocols for Building and Producing High Diversity Peptide Phage Display Libraries. Methods Mol Biol 2024; 2793:3-19. [PMID: 38526720 DOI: 10.1007/978-1-0716-3798-2_1] [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] [Indexed: 03/27/2024]
Abstract
Phage display is an important technology to study protein-protein interaction and protein evolution, with applications in basic science and applied biotechnology, such as drug discovery and the development of targeted therapies. However, in order to be successful during a phage display screening, it is paramount to have good phage libraries. Here, we described detailed procedures to generate peptide phage display libraries with high diversity and billions of transformants.
Collapse
Affiliation(s)
- Ricardo Jose Giordano
- Biochemistry Department, Institute of Chemistry, University of Sao Paulo, Sao Paulo, Brazil.
| | - Lilian Costa Alecrim
- Biochemistry Department, Institute of Chemistry, University of Sao Paulo, Sao Paulo, Brazil
| |
Collapse
|
8
|
Macias ALD, Alecrim LC, Almeida FCL, Giordano RJ. Understanding the Structural Requirements of Peptide-Protein Interaction and Applications for Peptidomimetic Development. Methods Mol Biol 2024; 2793:65-82. [PMID: 38526724 DOI: 10.1007/978-1-0716-3798-2_5] [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] [Indexed: 03/27/2024]
Abstract
Protein-protein interaction is at the heart of most biological processes, and small peptides that bind to protein binding sites are resourceful tools to explore and understand the structural requirements for these interactions. In that sense, phage display is a well-suited technology to study protein-protein interactions, as it allows for unbiased screening of billions of peptides in search for those that interact with a protein binding domain. Here, we will illustrate how two distinct but complementary approaches, phage display and nuclear magnetic resonance (NMR), can be utilized to unveil structural details of peptide-protein interaction. Finally, knowledge derived from phage mutagenesis and NMR studies can be streamlined for quick peptidomimetic design and synthesis using the retroinversion approach to validate using in vitro and in vivo assays the therapeutic potential of peptides identified by phage display.
Collapse
Affiliation(s)
| | - Lilian Costa Alecrim
- Biochemistry Department, Institute of Chemistry, University of Sao Paulo, Sao Paulo, Brazil
| | - Fabio C L Almeida
- Institute of Medical Biochemistry, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Ricardo Jose Giordano
- Biochemistry Department, Institute of Chemistry, University of Sao Paulo, Sao Paulo, Brazil.
| |
Collapse
|
9
|
Jin X, Ou Z, Zhang G, Shi R, Yang J, Liu W, Luo G, Deng J, Wang W. A CO-mediated photothermal therapy to kill drug-resistant bacteria and minimize thermal injury for infected diabetic wound healing. Biomater Sci 2023; 11:6236-6251. [PMID: 37531204 DOI: 10.1039/d3bm00774j] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/03/2023]
Abstract
With an increasing proportion of drug-resistant bacteria, photothermal therapy (PTT) is a promising alternative to antibiotic treatment for infected diabetic skin ulcers. However, the inevitable thermal damage to the tissues restricts its clinical practice. Carbon monoxide (CO), as a bioactive gas molecule, can selectively inhibit bacterial growth and promote tissue regeneration, which may be coordinated with PTT for drug-resistant bacteria killing and tissue protection. Herein, a CO-mediated PTT agent (CO@mPDA) was engineered by loading manganese carbonyl groups into mesoporous polydopamine (mPDA) nanoparticles via coordination interactions between the metal center and a catechol group. Compared to the traditional PTT, the CO-mediated PTT increases the inhibition ratio of the drug-resistant bacteria both in vitro and in diabetic wound beds by selectively inhibiting the co-chaperone of the heat shock protein 90 kDa (Hsp90), and lowers the heat resistance of the bacteria rather than the mammalian tissues. Meanwhile, the tissue-protective proteins, such as Hsp90 and vimentin (Vim), are upregulated via the WNT and PI3K-Akt pathways to reduce thermal injury, especially with a laser with a high-power density. The CO-mediated PTT unified the bacterial killing with tissue protection, which offers a promising concept to improve PTT efficiency and minimize the side-effects of PTT when treating infected skin wounds.
Collapse
Affiliation(s)
- Xin Jin
- Tianjin Key Laboratory of Composite and Functional Materials, School of Materials Science and Engineering, Tianjin, University, Tianjin 300350, China
| | - Zelin Ou
- Department of Nephrology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325035, China.
- Institute of Burn Research, State Key Lab of Trauma, Burn, and Combined Injury, Chongqing Key Laboratory for Disease Proteomics, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China.
| | - Guowei Zhang
- Institute of Burn Research, State Key Lab of Trauma, Burn, and Combined Injury, Chongqing Key Laboratory for Disease Proteomics, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China.
| | - Rong Shi
- Institute of Burn Research, State Key Lab of Trauma, Burn, and Combined Injury, Chongqing Key Laboratory for Disease Proteomics, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China.
| | - Jumin Yang
- Tianjin Key Laboratory of Composite and Functional Materials, School of Materials Science and Engineering, Tianjin, University, Tianjin 300350, China
| | - Wenguang Liu
- Tianjin Key Laboratory of Composite and Functional Materials, School of Materials Science and Engineering, Tianjin, University, Tianjin 300350, China
| | - Gaoxing Luo
- Department of Nephrology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325035, China.
- Institute of Burn Research, State Key Lab of Trauma, Burn, and Combined Injury, Chongqing Key Laboratory for Disease Proteomics, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China.
| | - Jun Deng
- Institute of Burn Research, State Key Lab of Trauma, Burn, and Combined Injury, Chongqing Key Laboratory for Disease Proteomics, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China.
| | - Wei Wang
- ZJU-Hangzhou Global Scientific and Technological Innovation Center, Hangzhou, Zhejiang 311215, China.
- College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310027, China
| |
Collapse
|
10
|
Namjoo M, Ghafouri H, Assareh E, Aref AR, Mostafavi E, Hamrahi Mohsen A, Balalaie S, Broussy S, Asghari SM. A VEGFB-Based Peptidomimetic Inhibits VEGFR2-Mediated PI3K/Akt/mTOR and PLCγ/ERK Signaling and Elicits Apoptotic, Antiangiogenic, and Antitumor Activities. Pharmaceuticals (Basel) 2023; 16:906. [PMID: 37375853 DOI: 10.3390/ph16060906] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Revised: 06/09/2023] [Accepted: 06/13/2023] [Indexed: 06/29/2023] Open
Abstract
Vascular endothelial growth factor receptor 2 (VEGFR2) mediates VEGFA signaling mainly through the PI3K/AKT/mTOR and PLCγ/ERK1/2 pathways. Here we unveil a peptidomimetic (VGB3) based on the interaction between VEGFB and VEGFR1 that unexpectedly binds and neutralizes VEGFR2. Investigation of the cyclic and linear structures of VGB3 (named C-VGB3 and L-VGB3, respectively) using receptor binding and cell proliferation assays, molecular docking, and evaluation of antiangiogenic and antitumor activities in the 4T1 mouse mammary carcinoma tumor (MCT) model showed that loop formation is essential for peptide functionality. C-VGB3 inhibited proliferation and tubulogenesis of human umbilical vein endothelial cells (HUVECs), accounting for the abrogation of VEGFR2, p-VEGFR2 and, subsequently, PI3K/AKT/mTOR and PLCγ/ERK1/2 pathways. In 4T1 MCT cells, C-VGB3 inhibited cell proliferation, VEGFR2 expression and phosphorylation, the PI3K/AKT/mTOR pathway, FAK/Paxillin, and the epithelial-to-mesenchymal transition cascade. The apoptotic effects of C-VGB3 on HUVE and 4T1 MCT cells were inferred from annexin-PI and TUNEL staining and activation of P53, caspase-3, caspase-7, and PARP1, which mechanistically occurred through the intrinsic pathway mediated by Bcl2 family members, cytochrome c, Apaf-1 and caspase-9, and extrinsic pathway via death receptors and caspase-8. These data indicate that binding regions shared by VEGF family members may be important in developing novel pan-VEGFR inhibitors that are highly relevant in the pathogenesis of angiogenesis-related diseases.
Collapse
Affiliation(s)
- Mohadeseh Namjoo
- Department of Biology, University Campus II, University of Guilan, Rasht P.O. Box 14155-6619, Iran
| | - Hossein Ghafouri
- Department of Biology, Faculty of Sciences, University of Guilan, Rasht P.O. Box 14155-6619, Iran
| | - Elham Assareh
- Department of Biology, Faculty of Sciences, University of Guilan, Rasht P.O. Box 14155-6619, Iran
| | - Amir Reza Aref
- Belfer Center for Applied Cancer Science, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Ebrahim Mostafavi
- Department of Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Ali Hamrahi Mohsen
- Institute of Biochemistry and Biophysics (IBB), University of Tehran, Tehran P.O. Box 1841, Iran
| | - Saeed Balalaie
- Peptide Chemistry Research Center, K. N. Toosi University of Technology, Tehran P.O. Box 1841, Iran
| | - Sylvain Broussy
- CiTCoM, UMR CNRS 8038, U1268 INSERM, UFR de Pharmacie, Faculté de Santé, Université Paris Cité, 75006 Paris, France
| | - S Mohsen Asghari
- Institute of Biochemistry and Biophysics (IBB), University of Tehran, Tehran P.O. Box 1841, Iran
| |
Collapse
|
11
|
Inague A, Alecrim LC, Monteiro JS, Yoshinaga MY, Setubal JC, Miyamoto S, Giordano RJ. Oxygen-induced pathological angiogenesis promotes intense lipid synthesis and remodeling in the retina. iScience 2023; 26:106777. [PMID: 37213234 PMCID: PMC10199268 DOI: 10.1016/j.isci.2023.106777] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 02/26/2023] [Accepted: 04/25/2023] [Indexed: 05/23/2023] Open
Abstract
The retina is a notable tissue with high metabolic needs which relies on specialized vascular networks to protect the neural retina while maintaining constant supplies of oxygen, nutrients, and dietary essential fatty acids. Here we analyzed the lipidome of the mouse retina under healthy and pathological angiogenesis using the oxygen-induced retinopathy model. By matching lipid profiles to changes in mRNA transcriptome, we identified a lipid signature showing that pathological angiogenesis leads to intense lipid remodeling favoring pathways for neutral lipid synthesis, cholesterol import/export, and lipid droplet formation. Noteworthy, it also shows profound changes in pathways for long-chain fatty acid production, vital for retina homeostasis. The net result is accumulation of large quantities of mead acid, a marker of essential fatty acid deficiency, and a potential marker for retinopathy severity. Thus, our lipid signature might contribute to better understand diseases of the retina that lead to vision impairment or blindness.
Collapse
Affiliation(s)
- Alex Inague
- Biochemistry Department, Institute of Chemistry, University of Sao Paulo, Sao Paulo, SP 05508-000, Brazil
| | - Lilian Costa Alecrim
- Biochemistry Department, Institute of Chemistry, University of Sao Paulo, Sao Paulo, SP 05508-000, Brazil
| | - Jhonatas Sirino Monteiro
- Biochemistry Department, Institute of Chemistry, University of Sao Paulo, Sao Paulo, SP 05508-000, Brazil
| | - Marcos Yukio Yoshinaga
- Biochemistry Department, Institute of Chemistry, University of Sao Paulo, Sao Paulo, SP 05508-000, Brazil
| | - João Carlos Setubal
- Biochemistry Department, Institute of Chemistry, University of Sao Paulo, Sao Paulo, SP 05508-000, Brazil
| | - Sayuri Miyamoto
- Biochemistry Department, Institute of Chemistry, University of Sao Paulo, Sao Paulo, SP 05508-000, Brazil
- Corresponding author
| | - Ricardo José Giordano
- Biochemistry Department, Institute of Chemistry, University of Sao Paulo, Sao Paulo, SP 05508-000, Brazil
- Corresponding author
| |
Collapse
|
12
|
Antiangiogenic potential of small polypeptide sequences: In vivo assays, cytotoxicity, synthetic approaches and influence of C-terminal carboxyamidation. J Mol Struct 2022. [DOI: 10.1016/j.molstruc.2022.133493] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
|
13
|
Peng Q, Sha K, Liu Y, Chen M, Xu S, Xie H, Deng Z, Li J. mTORC1-Mediated Angiogenesis is Required for the Development of Rosacea. Front Cell Dev Biol 2022; 9:751785. [PMID: 34993194 PMCID: PMC8724421 DOI: 10.3389/fcell.2021.751785] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Accepted: 12/08/2021] [Indexed: 12/14/2022] Open
Abstract
Although multiple evidences suggest that angiogenesis is associated with the pathophysiology of rosacea, its role is still in debate. Here, we showed that angiogenesis was enhanced in skin lesions of both rosacea patients and LL37-induced rosacea-like mice. Inhibition of angiogenesis alleviated LL37-induced rosacea-like features in mice. Mechanistically, we showed that mTORC1 was activated in the endothelial cells of the lesional skin from rosacea patients and LL37-induced rosacea-like mouse model. Inhibition of mTORC1 decreased angiogenesis and blocked the development of rosacea in mice. On the contrary, hyperactivation of mTORC1 increased angiogenesis and exacerbated rosacea-like phenotypes. Our in vitro results further demonstrated that inhibition of mTORC1 signaling significantly declined LL37-induced tube formation of human endothelial cells. Taken together, our findings revealed that mTORC1-mediated angiogenesis responding to LL37 might be essential for the development of rosacea and targeting angiogenesis might be a novel potential therapy.
Collapse
Affiliation(s)
- Qinqin Peng
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China.,Hunan Key Laboratary of Aging Biology, Xiangya Hospital, Central South University, Changsha, China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China.,Key Laboratory of Molecular Radiation Oncology of Hunan Province, Changsha, China
| | - Ke Sha
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China.,Hunan Key Laboratary of Aging Biology, Xiangya Hospital, Central South University, Changsha, China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China.,Key Laboratory of Molecular Radiation Oncology of Hunan Province, Changsha, China
| | - Yingzi Liu
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China.,Hunan Key Laboratary of Aging Biology, Xiangya Hospital, Central South University, Changsha, China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China.,Key Laboratory of Molecular Radiation Oncology of Hunan Province, Changsha, China
| | - Mengting Chen
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China.,Hunan Key Laboratary of Aging Biology, Xiangya Hospital, Central South University, Changsha, China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China.,Key Laboratory of Molecular Radiation Oncology of Hunan Province, Changsha, China
| | - San Xu
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China.,Hunan Key Laboratary of Aging Biology, Xiangya Hospital, Central South University, Changsha, China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China.,Key Laboratory of Molecular Radiation Oncology of Hunan Province, Changsha, China
| | - Hongfu Xie
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China.,Hunan Key Laboratary of Aging Biology, Xiangya Hospital, Central South University, Changsha, China.,Key Laboratory of Molecular Radiation Oncology of Hunan Province, Changsha, China.,Key Laboratory of Organ Injury, Aging and Regenerative Medicine of Hunan Province, Central South University, Changsha, China
| | - Zhili Deng
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China.,Hunan Key Laboratary of Aging Biology, Xiangya Hospital, Central South University, Changsha, China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China.,Key Laboratory of Molecular Radiation Oncology of Hunan Province, Changsha, China.,Key Laboratory of Organ Injury, Aging and Regenerative Medicine of Hunan Province, Central South University, Changsha, China
| | - Ji Li
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China.,Hunan Key Laboratary of Aging Biology, Xiangya Hospital, Central South University, Changsha, China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China.,Key Laboratory of Molecular Radiation Oncology of Hunan Province, Changsha, China.,Key Laboratory of Organ Injury, Aging and Regenerative Medicine of Hunan Province, Central South University, Changsha, China
| |
Collapse
|
14
|
Emerging Importance of Tyrosine Kinase Inhibitors against Cancer: Quo Vadis to Cure? Int J Mol Sci 2021; 22:ijms222111659. [PMID: 34769090 PMCID: PMC8584061 DOI: 10.3390/ijms222111659] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2021] [Revised: 10/24/2021] [Accepted: 10/24/2021] [Indexed: 12/12/2022] Open
Abstract
GLOBOCAN 2020 estimated more than 19.3 million new cases, and about 10 million patients were deceased from cancer in 2020. Clinical manifestations showed that several growth factor receptors consisting of transmembrane and cytoplasmic tyrosine kinase (TK) domains play a vital role in cancer progression. Receptor tyrosine kinases (RTKs) are crucial intermediaries of the several cellular pathways and carcinogenesis that directly affect the prognosis and survival of higher tumor grade patients. Tyrosine kinase inhibitors (TKIs) are efficacious drugs for targeted therapy of various cancers. Therefore, RTKs have become a promising therapeutic target to cure cancer. A recent report shows that TKIs are vital mediators of signal transduction and cancer cell proliferation, angiogenesis, and apoptosis. In this review, we discuss the structure and function of RTKs to explore their prime role in cancer therapy. Various TKIs have been developed to date that contribute a lot to treating several types of cancer. These TKI based anticancer drug molecules are also discussed in detail, incorporating their therapeutic efficacy, mechanism of action, and side effects. Additionally, this article focuses on TKIs which are running in the clinical trial and pre-clinical studies. Further, to gain insight into the pathophysiological mechanism of TKIs, we also reviewed the impact of RTK resistance on TKI clinical drugs along with their mechanistic acquired resistance in different cancer types.
Collapse
|
15
|
Design and In Silico Evaluation of a Novel Cyclic Disulfide-Rich anti-VEGF Peptide as a Potential Antiangiogenic Drug. Int J Pept Res Ther 2021. [DOI: 10.1007/s10989-021-10250-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
|
16
|
Ngema LM, Adeyemi SA, Marimuthu T, Choonara YE. A review on engineered magnetic nanoparticles in Non-Small-Cell lung carcinoma targeted therapy. Int J Pharm 2021; 606:120870. [PMID: 34245844 DOI: 10.1016/j.ijpharm.2021.120870] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Revised: 06/25/2021] [Accepted: 07/05/2021] [Indexed: 02/07/2023]
Abstract
There are growing appeals forthe design of efficacious treatment options for non-small-cell lung carcinoma (NSCLC) as it accrues to ~ 85% cases of lung cancer. Although platinum-based doublet chemotherapy has been the main therapeutic intervention in NSCLC management, this leads to myriad of problems including intolerability to the doublet regimens and detrimental side effects due to high doses. A new approach is therefore needed and warrants the design of targeted drug delivery systems that can halt tumor proliferation and metastasis by targeting key molecules, while exhibiting minimal side effects and toxicity. This review aims to explore the rational design of magnetic nanoparticles for the development of tumor-targeting systems for NSCLC. In the review, we explore the anticancer merits of conjugated linoleic acid (CLA) and provide a concise incursion into its application for the invention of functionalized magnetic nanoparticles in the targeted treatment of NSCLC. Recent nanoparticle-based targeted chemotherapies for targeting angiogenesis biomarkers in NSCLC will also be reviewed to further highlight versatility of magnetic nanoparticles. These developments through molecular tuning at the nanoscale and supported by comprehensive pre-clinical studies could lead to the establishment of precise nanosystems for tumor-homing cancer therapy.
Collapse
Affiliation(s)
- Lindokuhle M Ngema
- Wits Advanced Drug Delivery Platform Research Unit, Department of Pharmacy and Pharmacology, School of Therapeutic Sciences, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, 7 York Road, Parktown 2193, South Africa
| | - Samson A Adeyemi
- Wits Advanced Drug Delivery Platform Research Unit, Department of Pharmacy and Pharmacology, School of Therapeutic Sciences, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, 7 York Road, Parktown 2193, South Africa
| | - Thashree Marimuthu
- Wits Advanced Drug Delivery Platform Research Unit, Department of Pharmacy and Pharmacology, School of Therapeutic Sciences, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, 7 York Road, Parktown 2193, South Africa
| | - Yahya E Choonara
- Wits Advanced Drug Delivery Platform Research Unit, Department of Pharmacy and Pharmacology, School of Therapeutic Sciences, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, 7 York Road, Parktown 2193, South Africa.
| |
Collapse
|
17
|
Anti-Angiogenic Property of Free Human Oligosaccharides. Biomolecules 2021; 11:biom11060775. [PMID: 34064180 PMCID: PMC8224327 DOI: 10.3390/biom11060775] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Revised: 05/18/2021] [Accepted: 05/19/2021] [Indexed: 12/29/2022] Open
Abstract
Angiogenesis, a fundamental process in human physiology and pathology, has attracted considerable attention owing to its potential as a therapeutic strategy. Vascular endothelial growth factor (VEGF) and its receptor (VEGFR) are deemed major mediators of angiogenesis. To date, inhibition of the VEGF-A/VEGFR-2 axis has been an effective strategy employed in the development of anticancer drugs. However, some limitations, such as low efficacy and side effects, need to be addressed. Several drug candidates have been discovered, including small molecule compounds, recombinant proteins, and oligosaccharides. In this review, we focus on human oligosaccharides as modulators of angiogenesis. In particular, sialylated human milk oligosaccharides (HMOs) play a significant role in the inhibition of VEGFR-2-mediated angiogenesis. We discuss the structural features concerning the interaction between sialylated HMOs and VEGFR-2 as a molecular mechanism of anti-angiogenesis modulation and its effectiveness in vivo experiments. In the current state, extensive clinical trials are required to develop a novel VEGFR-2 inhibitor from sialylated HMOs.
Collapse
|
18
|
Teixeira AAR, Carnero LR, Kuramoto A, Tang FHF, Gomes CH, Pereira NB, de Oliveira LC, Garrini R, Monteiro JS, Setubal JC, Sabino EC, Pasqualini R, Colli W, Arap W, Alves MJM, Cunha-Neto E, Giordano RJ. A refined genome phage display methodology delineates the human antibody response in patients with Chagas disease. iScience 2021; 24:102540. [PMID: 34142048 PMCID: PMC8185243 DOI: 10.1016/j.isci.2021.102540] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Revised: 03/30/2021] [Accepted: 05/12/2021] [Indexed: 12/23/2022] Open
Abstract
Large-scale mapping of antigens and epitopes is pivotal for developing immunotherapies but challenging, especially for eukaryotic pathogens, owing to their large genomes. Here, we developed an integrated platform for genome phage display (gPhage) to show that unbiased libraries of the eukaryotic parasite Trypanosoma cruzi enable the identification of thousands of antigens recognized by serum samples from patients with Chagas disease. Because most of these antigens are hypothetical proteins, gPhage provides evidence of their expression during infection. We built and validated a comprehensive map of Chagas disease antibody response to show how linear and putative conformation epitopes, many rich in repetitive elements, allow the parasite to evade a buildup of neutralizing antibodies directed against protein domains that mediate infection pathogenesis. Thus, the gPhage platform is a reproducible and effective tool for rapid simultaneous identification of epitopes and antigens, not only in Chagas disease but perhaps also in globally emerging/reemerging acute pathogens. Genomic shotgun phage display (gPhage) of eukaryotes is feasible and promising. gPhage allows rapid antigen ID and epitope mapping, including 3D structures. Conformation epitopes can be identified and validated by using the gPhage platform. Most Chagas disease antigens are hypothetical proteins rich in repetitive elements.
Collapse
Affiliation(s)
- André Azevedo Reis Teixeira
- Department of Biochemistry, Institute of Chemistry, University of São Paulo, São Paulo, SP, 05508-000, Brazil
| | - Luis Rodriguez Carnero
- Department of Biochemistry, Institute of Chemistry, University of São Paulo, São Paulo, SP, 05508-000, Brazil
| | - Andréia Kuramoto
- Heart Institute (InCor), University of São Paulo School of Medicine, São Paulo, SP, 05403-000, Brazil
| | - Fenny Hui Fen Tang
- Department of Biochemistry, Institute of Chemistry, University of São Paulo, São Paulo, SP, 05508-000, Brazil.,Division of Cancer Biology, Department of Radiation Oncology, Rutgers New Jersey Medical School, Newark, NJ 07103, USA
| | - Carlos Hernique Gomes
- Department of Biochemistry, Institute of Chemistry, University of São Paulo, São Paulo, SP, 05508-000, Brazil
| | - Natalia Bueno Pereira
- Heart Institute (InCor), University of São Paulo School of Medicine, São Paulo, SP, 05403-000, Brazil
| | - Léa Campos de Oliveira
- Institute of Tropical Medicine, University of São Paulo School of Medicine, São Paulo, SP, 05403-000, Brazil
| | - Regina Garrini
- Institute of Tropical Medicine, University of São Paulo School of Medicine, São Paulo, SP, 05403-000, Brazil
| | - Jhonatas Sirino Monteiro
- Department of Biochemistry, Institute of Chemistry, University of São Paulo, São Paulo, SP, 05508-000, Brazil
| | - João Carlos Setubal
- Department of Biochemistry, Institute of Chemistry, University of São Paulo, São Paulo, SP, 05508-000, Brazil
| | - Ester Cerdeira Sabino
- Institute of Tropical Medicine, University of São Paulo School of Medicine, São Paulo, SP, 05403-000, Brazil
| | - Renata Pasqualini
- Rutgers Cancer Institute of New Jersey, Newark, NJ 07103, USA.,Division of Cancer Biology, Department of Radiation Oncology, Rutgers New Jersey Medical School, Newark, NJ 07103, USA
| | - Walter Colli
- Department of Biochemistry, Institute of Chemistry, University of São Paulo, São Paulo, SP, 05508-000, Brazil
| | - Wadih Arap
- Rutgers Cancer Institute of New Jersey, Newark, NJ 07103, USA.,Division of Hematology/Oncology, Department of Medicine, Rutgers New Jersey Medical School, Newark, NJ 07103, USA
| | - Maria Júlia Manso Alves
- Department of Biochemistry, Institute of Chemistry, University of São Paulo, São Paulo, SP, 05508-000, Brazil
| | - Edécio Cunha-Neto
- Heart Institute (InCor), University of São Paulo School of Medicine, São Paulo, SP, 05403-000, Brazil.,Division of Clinical Immunology and Allergy, University of São Paulo School of Medicine, São Paulo, SP 01246-903, Brazil.,Institute for Investigation in Immunology (iii), INCT, São Paulo, SP, Brazil
| | - Ricardo José Giordano
- Department of Biochemistry, Institute of Chemistry, University of São Paulo, São Paulo, SP, 05508-000, Brazil.,Institute for Investigation in Immunology (iii), INCT, São Paulo, SP, Brazil
| |
Collapse
|
19
|
Wen S, Zhang K, Li Y, Fan J, Ziming C, Zhang J, Wang H, Wang L. A self-assembling peptide targeting VEGF receptors to inhibit angiogenesis. CHINESE CHEM LETT 2020. [DOI: 10.1016/j.cclet.2020.03.077] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
20
|
Guarischi-Sousa R, Monteiro JS, Alecrim LC, Michaloski JS, Cardeal LB, Ferreira EN, Carraro DM, Nunes DN, Dias-Neto E, Reimand J, Boutros PC, Setubal JC, Giordano RJ. A transcriptome-based signature of pathological angiogenesis predicts breast cancer patient survival. PLoS Genet 2019; 15:e1008482. [PMID: 31846472 PMCID: PMC6917213 DOI: 10.1371/journal.pgen.1008482] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2019] [Accepted: 10/15/2019] [Indexed: 12/30/2022] Open
Abstract
The specific genes and molecules that drive physiological angiogenesis differ from those involved in pathological angiogenesis, suggesting distinct mechanisms for these seemingly related processes. Unveiling genes and pathways preferentially associated with pathologic angiogenesis is key to understanding its mechanisms, thereby facilitating development of novel approaches to managing angiogenesis-dependent diseases. To better understand these different processes, we elucidated the transcriptome of the mouse retina in the well-accepted oxygen-induced retinopathy (OIR) model of pathological angiogenesis. We identified 153 genes changed between normal and OIR retinas, which represent a molecular signature relevant to other angiogenesis-dependent processes such as cancer. These genes robustly predict the survival of breast cancer patients, which was validated in an independent 1,000-patient test cohort (40% difference in 15-year survival; p = 2.56 x 10-21). These results suggest that the OIR model reveals key genes involved in pathological angiogenesis, and these may find important applications in stratifying tumors for treatment intensification or for angiogenesis-targeted therapies.
Collapse
Affiliation(s)
- Rodrigo Guarischi-Sousa
- Biochemistry Department, Institute of Chemistry, University of São Paulo, São Paulo, Brazil
- Computational Biology Program, Ontario Institute for Cancer Research, Toronto, Ontario, Canada
| | - Jhonatas S. Monteiro
- Biochemistry Department, Institute of Chemistry, University of São Paulo, São Paulo, Brazil
| | - Lilian C. Alecrim
- Biochemistry Department, Institute of Chemistry, University of São Paulo, São Paulo, Brazil
| | - Jussara S. Michaloski
- Biochemistry Department, Institute of Chemistry, University of São Paulo, São Paulo, Brazil
| | - Laura B. Cardeal
- Biochemistry Department, Institute of Chemistry, University of São Paulo, São Paulo, Brazil
| | - Elisa N. Ferreira
- International Research Center (CIPE) A.C. Camargo Cancer Center, São Paulo, SP, Brazil
| | - Dirce M. Carraro
- International Research Center (CIPE) A.C. Camargo Cancer Center, São Paulo, SP, Brazil
| | - Diana N. Nunes
- International Research Center (CIPE) A.C. Camargo Cancer Center, São Paulo, SP, Brazil
| | - Emmanuel Dias-Neto
- International Research Center (CIPE) A.C. Camargo Cancer Center, São Paulo, SP, Brazil
- Laboratory of Neurosciences (LIM27), Institute & Department of Psychiatry, University of São Paulo, São Paulo, Brazil
| | - Jüri Reimand
- Computational Biology Program, Ontario Institute for Cancer Research, Toronto, Ontario, Canada
- Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada
| | - Paul C. Boutros
- Department of Human Genetics, University of California Los Angeles (UCLA), Los Angeles, CA, United States of America
- * E-mail: (PCB); (JCS); (RJG)
| | - João C. Setubal
- Biochemistry Department, Institute of Chemistry, University of São Paulo, São Paulo, Brazil
- * E-mail: (PCB); (JCS); (RJG)
| | - Ricardo J. Giordano
- Biochemistry Department, Institute of Chemistry, University of São Paulo, São Paulo, Brazil
- * E-mail: (PCB); (JCS); (RJG)
| |
Collapse
|
21
|
Khodabakhsh F, Muyldermans S, Behdani M, Kazemi-Lomedasht F. Liposomal delivery of vascular endothelial growth factor/receptors and their inhibitors. J Drug Target 2019; 28:379-385. [DOI: 10.1080/1061186x.2019.1693578] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Affiliation(s)
- Farnaz Khodabakhsh
- Department of Genetics and Advanced Medical Technology, Medical Biotechnology Research Center, Faculty of Medicine, AJA University of Medical Sciences, Tehran, Iran
| | - Serge Muyldermans
- Laboratory of Cellular and Molecular Immunology, Vrije Universiteit Brussel, Brussels, Belgium
| | - Mahdi Behdani
- Biotechnology Research Center, Venom and Biotherapeutics Molecules Laboratory, Pasteur Institute of Iran, Tehran, Iran
| | - Fatemeh Kazemi-Lomedasht
- Biotechnology Research Center, Venom and Biotherapeutics Molecules Laboratory, Pasteur Institute of Iran, Tehran, Iran
| |
Collapse
|
22
|
Faulin TDES, Kazuma SM, Tripodi GL, Cavalcante MF, Wakasuqui F, Oliveira CLP, Degenhardt MFDS, Michaloski J, Giordano RJ, Ketelhuth DFJ, Abdalla DSP. Proinflammatory Action of a New Electronegative Low-Density Lipoprotein Epitope. Biomolecules 2019; 9:biom9080386. [PMID: 31434316 PMCID: PMC6723646 DOI: 10.3390/biom9080386] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2019] [Revised: 08/03/2019] [Accepted: 08/13/2019] [Indexed: 01/02/2023] Open
Abstract
The electronegative low-density lipoprotein, LDL (-), is an endogenously modified LDL subfraction with cytotoxic and proinflammatory actions on endothelial cells, monocytes, and macrophages contributing to the progression of atherosclerosis. In this study, epitopes of LDL (-) were mapped using a phage display library of peptides and monoclonal antibodies reactive to this modified lipoprotein. Two different peptide libraries (X6 and CX8C for 6- and 8-amino acid-long peptides, respectively) were used in the mapping. Among all tested peptides, two circular peptides, P1A3 and P2C7, were selected based on their high affinities for the monoclonal antibodies. Small-angle X-ray scattering analysis confirmed their structures as circular rings. P1A3 or P2C7 were quickly internalized by bone marrow-derived murine macrophages as shown by confocal microscopy. P2C7 increased the expression of TNFα, IL-1 β and iNOS as well as the secretion of TNFα, CCL2, and nitric oxide by murine macrophages, similar to the responses induced by LDL (-), although less intense. In contrast, P1A3 did not show pro-inflammatory effects. We identified a mimetic epitope associated with LDL (-), the P2C7 circular peptide, that activates macrophages. Our data suggest that this conformational epitope represents an important danger-associated molecular pattern of LDL (-) that triggers proinflammatory responses.
Collapse
Affiliation(s)
- Tanize do Espirito Santo Faulin
- Department of Clinical and Toxicological Analyses, Faculty of Pharmaceutical Sciences, University of Sao Paulo, Sao Paulo 05508-000, Brazil
| | - Soraya Megumi Kazuma
- Department of Clinical and Toxicological Analyses, Faculty of Pharmaceutical Sciences, University of Sao Paulo, Sao Paulo 05508-000, Brazil
| | - Gustavo Luis Tripodi
- Department of Clinical and Toxicological Analyses, Faculty of Pharmaceutical Sciences, University of Sao Paulo, Sao Paulo 05508-000, Brazil
| | - Marcela Frota Cavalcante
- Department of Clinical and Toxicological Analyses, Faculty of Pharmaceutical Sciences, University of Sao Paulo, Sao Paulo 05508-000, Brazil
| | - Felipe Wakasuqui
- Department of Clinical and Toxicological Analyses, Faculty of Pharmaceutical Sciences, University of Sao Paulo, Sao Paulo 05508-000, Brazil
| | | | | | - Jussara Michaloski
- Department of Biochemistry, Institute of Chemistry, University of Sao Paulo, Sao Paulo 05508-000, SP, Brazil
| | - Ricardo José Giordano
- Department of Biochemistry, Institute of Chemistry, University of Sao Paulo, Sao Paulo 05508-000, SP, Brazil
| | - Daniel Francisco Jacon Ketelhuth
- Centre for Molecular Medicine, Department of Medicine, Karolinska University Hospital, Karolinska Institute, 17164 Stockholm, Sweden
- Department of Cardiovascular and Renal Research, Institute for Molecular Medicine, University of Southern Denmark (SDU), 5000 Odense, Denmark
| | - Dulcineia Saes Parra Abdalla
- Department of Clinical and Toxicological Analyses, Faculty of Pharmaceutical Sciences, University of Sao Paulo, Sao Paulo 05508-000, Brazil.
| |
Collapse
|
23
|
Carneiro-Lobo TC, Scalabrini LC, Magalhães LDS, Cardeal LB, Rodrigues FS, Dos Santos EO, Baldwin AS, Levantini E, Giordano RJ, Bassères DS. IKKβ targeting reduces KRAS-induced lung cancer angiogenesis in vitro and in vivo: A potential anti-angiogenic therapeutic target. Lung Cancer 2019; 130:169-178. [PMID: 30885340 DOI: 10.1016/j.lungcan.2019.02.027] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2018] [Revised: 02/14/2019] [Accepted: 02/25/2019] [Indexed: 12/15/2022]
Abstract
OBJECTIVES The ability of tumor cells to drive angiogenesis is an important cancer hallmark that positively correlates with metastatic potential and poor prognosis. Therefore, targeting angiogenesis is a rational therapeutic approach and dissecting proangiogenic pathways is important, particularly for malignancies driven by oncogenic KRAS, which are widespread and lack effective targeted therapies. Based on published studies showing that oncogenic RAS promotes angiogenesis by upregulating the proangiogenic NF-κB target genes IL-8 and VEGF, that NF-κB activation by KRAS requires the IKKβ kinase, and that targeting IKKβ reduces KRAS-induced lung tumor growth in vivo, but has limited effects on cell growth in vitro, we hypothesized that IKKβ targeting would reduce lung tumor growth by inhibiting KRAS-induced angiogenesis. MATERIALS AND METHODS To test this hypothesis, we targeted IKKβ in KRAS-mutant lung cancer cell lines either by siRNA-mediated transfection or by treatment with Compound A (CmpdA), a highly specific IKKβ inhibitor, and used in vitro and in vivo assays to evaluate angiogenesis. RESULTS AND CONCLUSIONS Both pharmacological and siRNA-mediated IKKβ targeting in lung cells reduced expression and secretion of NF-κB-regulated proangiogenic factors IL-8 and VEGF. Moreover, conditioned media from IKKβ-targeted lung cells reduced human umbilical vein endothelial cell (HUVEC) migration, invasion and tube formation in vitro. Furthermore, siRNA-mediated IKKβ inhibition reduced xenograft tumor growth and vascularity in vivo. Finally, IKKβ inhibition also affects endothelial cell function in a cancer-independent manner, as IKKβ inhibition reduced pathological retinal angiogenesis in a mouse model of oxygen-induced retinopathy. Taken together, these results provide a novel mechanistic understanding of how the IKKβ pathway affects human lung tumorigenesis, indicating that IKKβ promotes KRAS-induced angiogenesis both by cancer cell-intrinsic and cancer cell-independent mechanisms, which strongly suggests IKKβ inhibition as a promising antiangiogenic approach to be explored for KRAS-induced lung cancer therapy.
Collapse
Affiliation(s)
| | | | | | - Laura B Cardeal
- Chemistry Institute, Department of Biochemistry, University of São Paulo, São Paulo, Brazil
| | - Felipe Silva Rodrigues
- Chemistry Institute, Department of Biochemistry, University of São Paulo, São Paulo, Brazil
| | | | - Albert S Baldwin
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Elena Levantini
- Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA; Institute of Biomedical Technologies, National Research Council (CNR), Pisa, Italy
| | - Ricardo J Giordano
- Chemistry Institute, Department of Biochemistry, University of São Paulo, São Paulo, Brazil
| | | |
Collapse
|
24
|
A ligand motif enables differential vascular targeting of endothelial junctions between brain and retina. Proc Natl Acad Sci U S A 2019; 116:2300-2305. [PMID: 30670660 DOI: 10.1073/pnas.1809483116] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Endothelial heterogeneity has important implications in health and disease. Molecular markers selectively expressed in the vasculature of different organs and tissues are currently being explored in targeted therapies with promising results in preclinical and clinical studies. Noteworthy is the role that combinatorial approaches such as phage display have had in identifying such markers by using phage as nanoparticles and surrogates for billions of different peptides, screening noninvasively the vascular lumen for binding sites. Here, we show that a new peptide motif that emerged from such combinatorial screening of the vasculature binds selectively to blood vessels in the brain in vivo but not to vessels in other organs. Peptides containing a conserved motif in which amino acids Phenylalanine-Arginine-Tryptophan (FRW) predominate could be visualized by transmission electron microscopy bound to the junctions between endothelial cells in all areas of the brain, including the optic nerve, but not in other barrier-containing tissues, such as intestines and testis. Remarkably, peptides containing the motif do not bind to vessels in the retina, implying an important molecular difference between these two vascular barriers. Furthermore, the peptide allows for in vivo imaging, demonstrating that new tools for studying and imaging the brain are likely to emerge from this motif.
Collapse
|
25
|
Sadremomtaz A, Mansouri K, Alemzadeh G, Safa M, Rastaghi AE, Asghari SM. Dual blockade of VEGFR1 and VEGFR2 by a novel peptide abrogates VEGF-driven angiogenesis, tumor growth, and metastasis through PI3K/AKT and MAPK/ERK1/2 pathway. Biochim Biophys Acta Gen Subj 2018; 1862:2688-2700. [DOI: 10.1016/j.bbagen.2018.08.013] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2018] [Revised: 08/02/2018] [Accepted: 08/09/2018] [Indexed: 12/14/2022]
|