1
|
Rahdari T, Ghafouri H, Ramezanpour S, Ardestani MS, Asghari SM. Design and Characterization of Peptide-Conjugated Solid Lipid Nanoparticles for Targeted MRI and SPECT Imaging of Breast Tumors. ACS OMEGA 2025; 10:17310-17326. [PMID: 40352495 PMCID: PMC12059910 DOI: 10.1021/acsomega.4c10153] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/07/2024] [Revised: 03/17/2025] [Accepted: 03/31/2025] [Indexed: 05/14/2025]
Abstract
Triple-negative breast cancer (TNBC) presents significant challenges due to its aggressive behavior and lack of targeted treatments. High-resolution imaging techniques and targeted nanoparticles offer potential solutions for early detection and monitoring of TNBC. In this study, we developed and characterized solid lipid nanoparticles (SLNs) conjugated with a C-peptide derived from endostatin to target integrin αvβ3, overexpressed in TNBC. These SLNs were loaded with superparamagnetic iron oxide nanoparticles (SPIONs) for enhanced magnetic resonance imaging (MRI) and radiolabeled with technetium-99m (99mTc) for single-photon emission computed tomography (SPECT), enabling dual-modality imaging. Extensive characterization of the nanoparticles was performed utilizing a variety of advanced techniques, including dynamic light scattering (DLS), differential scanning calorimetry (DSC), thermogravimetric analysis (TGA), X-ray diffraction (XRD), vibrating sample magnetometry (VSM), field-emission scanning electron microscopy (FE-SEM), and atomic force microscopy (AFM). This comprehensive analysis validated the successful synthesis and functionalization of the nanoparticles, along with their remarkable magnetic properties, while also revealing their distinct spherical morphology, optimal size, uniform distribution, and colloidal stability. The conjugation of C-peptide significantly enhanced the targeting efficiency in vitro, as evidenced by the MTT and receptor-binding assays in 4T1 cells using flow cytometry and MRI. In vivo studies using a 4T1 murine model demonstrated that peptide-conjugated SLNs accumulated in tumor tissues, providing superior contrast in MRI and enhanced tumor-specific localization in SPECT imaging. Biodistribution analysis confirmed reduced off-target accumulation, particularly in the liver, compared to nontargeted formulations. Collectively, C-peptide-conjugated SLNs provide a promising dual-modality imaging platform for TNBC, offering improved diagnostic accuracy and tumor targeting.
Collapse
Affiliation(s)
- Tahereh Rahdari
- Department
of Biology, Faculty of Sciences, University
of Guilan, 4199613776 Rasht, Iran
| | - Hossein Ghafouri
- Department
of Biology, Faculty of Sciences, University
of Guilan, 4199613776 Rasht, Iran
| | - Sorour Ramezanpour
- Department
of Chemistry, K. N. Toosi University of
Technology, 158754416 Tehran, Iran
| | - Mehdi Shafiee Ardestani
- Department
of Radiopharmacy, Faculty of Pharmacy, Tehran
University of Medical Sciences, 1461884513 Tehran, Iran
- Research
Center for Nuclear Medicine, Shariati Hospital, North Kargar Ave, 1411713135 Tehran, Iran
| | - S. Mohsen Asghari
- Institute
of Biochemistry and Biophysics, University
of Tehran, 1411713135 Tehran, Iran
| |
Collapse
|
2
|
Novel Anti-angiogenic Peptide Derived from Canstatin Induces Apoptosis In Vitro and In Vivo. Int J Pept Res Ther 2022. [DOI: 10.1007/s10989-022-10458-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/14/2022]
|
3
|
Chamani R, Taleqani MH, Imanpour A, Khatami M. New insights into short peptides derived from the collagen NC1 α1, α2, and α3 (IV) domains: An experimental and MD simulations study. BIOCHIMICA ET BIOPHYSICA ACTA. PROTEINS AND PROTEOMICS 2022; 1870:140769. [PMID: 35151893 DOI: 10.1016/j.bbapap.2022.140769] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Revised: 02/04/2022] [Accepted: 02/08/2022] [Indexed: 10/19/2022]
Abstract
Proteolytically cleavage of the collagen NC1 α1 to α3 (IV) domains leads to antiangiogenic proteins called Arresten, Canstatin, and Tumstatin, respectively. The research identified that the two overlapping peptides derived from Tumstatin are more effective than other fragments and amino acids L78, V82, and D84 are essential for their activity. In the present study, the efficacy of a nine amino acid peptide derived from Tumstatin (Tum), containing amino acids L78, V82, and D84 was compared to the corresponding sequence in Arresten (Ars) and Canstatin (Can) in vitro and in vivo. Moreover, CD spectroscopy, MD, and docking simulations were performed to evaluate the structure and the interaction of peptides to integrin αvβ3, respectively. Results demonstrated that peptides inhibit viability, migration, and tube formation in vitro, as well as the growth of tumor in vivo and Canstatin-derived peptide was more potent than others. CD measurement and DSSP calculation revealed that Can had more coil conformation. According to MD simulations, Can had more fluctuation, less intramolecular interactions, and less structural compactness compared to Tum and Ars. It can be assumed that amino acid variations lead to a more flexible and loose structure compared to the other peptides. The Canstatin-derived peptide interacts with the integrin αvβ3 extremely close to RGD binding site by the most negative binding energy and more interactions. In conclusion, we for the first time identified an active peptide derived from Canstatin and showed that the sequence affected structure and thereby interaction of peptide to its receptor.
Collapse
Affiliation(s)
- Reyhane Chamani
- Department of Biology, Faculty of Science, Yazd University, Yazd, Iran.
| | | | - Aylar Imanpour
- Department of Fisheries, Faculty of Natural Resources, Urmia University, Urmia, Iran
| | - Mehri Khatami
- Department of Biology, Faculty of Science, Yazd University, Yazd, Iran
| |
Collapse
|
4
|
Ji Y, Fan H, Yang M, Bai C, Yang W, Wang Z. Synergistic effect of baculovirus-mediated endostatin and angiostatin combined with gemcitabine in hepatocellular carcinoma. Biol Pharm Bull 2021; 45:309-315. [PMID: 34937830 DOI: 10.1248/bpb.b21-00857] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Anti-angiogenic gene therapy is a promising strategy in treating cancer. Endostatin and angiostatin are widely used in tumor anti-angiogenesis therapy. Our previous studies have shown that the BDS-hEA, a baculovirus long-term expressing the fusion protein of human endostatin and angiostatin, has a favorable effect in inhibiting the growth and angiogenesis of hepatocellular carcinoma. The purpose of this study was to further investigate its synergistic antitumor efficiency in combination with low-dose chemotherapeutic gemcitabine (GEM) on the subcutaneous hepatocellular carcinoma xenograft model in nude mice. The results showed that the combined group significantly inhibited (P<0.05 or P<0.01 or P<0.001) the growth of tumor weight and volume, reduced the expression of ki67 (cell proliferation marker), CD31 (angiogenic marker) and Matrix metalloproteinase 9 (MMP-9, tumor invasion and metastasis marker) and increased the apoptosis of tumor cells compared with the monotherapy and control groups, respectively. Synergistic index results showed that BDS-hEA combined with GEM had a synergistic effect in inhibiting tumor volume, proliferation, microvessel density, metastasis and promoting tumor apoptosis. Furthermore, there were no metastatic nodules and obvious pathological changes in liver tissue of the combined group, and the serum liver function indicators aspartate aminotransferase (AST), alanine aminotransferase (ALT), total bilirubin (T-BIL), alkaline phosphatase (ALP) and glutamyl transpeptidase (GGT) were significantly reduced (P<0.05 or P<0.01 or P<0.001) in the BDS-hEA or GEM groups compared with the control group. Notably, the combined therapy showed lower levels of liver function indicators than the GEM group. These data support the view that the combination of BDS-hEA and GEM has a synergistic anti-tumor properties and can reduce the damage of liver to certain extent.
Collapse
Affiliation(s)
- Yonggan Ji
- School of Pharmacy, Ningxia Medical University
| | - Hongli Fan
- School of Pharmacy, Ningxia Medical University
| | - Mengmeng Yang
- Laboratory Animal Center, Ningxia Medical University
| | | | - Wen Yang
- Laboratory Animal Center, Ningxia Medical University
| | - Zhisheng Wang
- School of Pharmacy, Ningxia Medical University.,Laboratory Animal Center, Ningxia Medical University
| |
Collapse
|
5
|
Chamani R, Soleimanjahi H, Asghari SM, Karimi H, Kianmehr Z, Ardestani SK. Re-engineering of the Immunosuppressive Tumor Microenvironment by Antiangiogenic Therapy. Int J Pept Res Ther 2020. [DOI: 10.1007/s10989-019-09860-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
6
|
Sadremomtaz A, Kobarfard F, Mansouri K, Mirzanejad L, Asghari SM. Suppression of migratory and metastatic pathways via blocking VEGFR1 and VEGFR2. J Recept Signal Transduct Res 2019; 38:432-441. [PMID: 30929546 DOI: 10.1080/10799893.2019.1567785] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
BACKGROUND Vascular endothelial growth factor (VEGF) A and B are endothelial cell mitogens whose ligation to VEGFR1/VEGFR2 drives tumor angiogenesis and metastasis, and epithelial-mesenchymal transition (EMT). Blockade of these signaling axes could be obtained by disturbing the interactions between VEGFA and/or VEGFB with VEGFR1 and/or VEGFR2. METHODS A 14-mer peptide (VGB) that recognizes both VEGFR1 and VEGFR2 were investigated for its inhibitory effects on the VEGF-induced proliferation and migration using MTT and scratch assay, respectively. Downstream signaling pathways were also assessed by quantitative estimation of gene and protein expression using real-time PCR and immunohistochemistry (IHC). RESULTS We investigated the inhibitory effects of VGB on downstream mediators of metastasis, including epithelial-cadherin (E-cadherin), matrix metalloprotease-9 (MMP-9), cancer myelocytomatosis (c-Myc), and nuclear factor-κβ (NF-κβ), and migration, comprising focal adhesion kinase (FAK) and its substrate Paxilin. VGB inhibited the VEGF-induced proliferation of human umbilical vein endothelial cells (HUVECs), 4T1 and U87 cells in a time- and dose-dependent manner and migration of HUVECs. Based on IHC analyses, treatment of 4T1 mammary carcinoma tumor with VGB led to the suppression of p-AKT, p-ERK1/2, MMP-9, NF-κβ, and activation of E-cadherin compared with PBS-treated controls. Moreover, quantitative real-time PCR analyses of VGB-treated tumors revealed the reduced expression level of FAK, Paxilin, NF-κβ, MMP-9, c-Myc, and increased expression level of E-cadherin compared to PBS-treated controls. CONCLUSIONS Our results demonstrated that simultaneous blockade of VEGFR1/VEGFR2 is an effective strategy to fight solid tumors by targeting a wider range of mediators involved in tumor angiogenesis, growth, and metastasis.
Collapse
Affiliation(s)
- Afsaneh Sadremomtaz
- a Department of Biology, Faculty of Sciences , University of Guilan , Rasht , Iran
| | - Farzad Kobarfard
- b Department of Medicinal Chemistry, School of Pharmacy , Phytochemistry Research Center, Shahid Beheshti University of Medical Sciences , Tehran , Iran
| | - Kamran Mansouri
- c Medical Biology Research Center, Kermanshah University of Medical Sciences , Kermanshah , Iran
| | - Laleh Mirzanejad
- a Department of Biology, Faculty of Sciences , University of Guilan , Rasht , Iran
| | - S Mohsen Asghari
- a Department of Biology, Faculty of Sciences , University of Guilan , Rasht , Iran
| |
Collapse
|
7
|
A cyclic peptide reproducing the α1 helix of VEGF-B binds to VEGFR-1 and VEGFR-2 and inhibits angiogenesis and tumor growth. Biochem J 2019; 476:645-663. [DOI: 10.1042/bcj20180823] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2018] [Revised: 01/26/2019] [Accepted: 01/29/2019] [Indexed: 11/17/2022]
Abstract
Abstract
Vascular endothelial growth factors (VEGFs) and their receptors (VEGFRs) are pivotal regulators of angiogenesis. The VEGF–VEGFR system is therefore an important target of anti-angiogenesis therapy. Based on the X-ray structure of VEGF-B/VEGFR-1 D2, we designed a cyclic peptide (known as VGB1) reproducing the α1 helix and its adjacent region to interfere with signaling through VEGFR-1. Unexpectedly, VGB1 bound VEGFR-2 in addition to VEGFR-1, leading to inhibition of VEGF-stimulated proliferation of human umbilical vein endothelial cells and 4T1 murine mammary carcinoma cells, which express VGEFR-1 and VEGFR-2, and U87 glioblastoma cells that mostly express VEGFR-2. VGB1 inhibited different aspects of angiogenesis, including proliferation, migration and tube formation of endothelial cells stimulated by VEGF-A through suppression of extracellular signal-regulated kinase 1/2 and AKT (Protein Kinase B) phosphorylation. In a murine 4T1 mammary carcinoma model, VGB1 caused regression of tumors without causing weight loss in association with impaired cell proliferation (decreased Ki67 expression) and angiogenesis (decreased CD31 and CD34 expression), and apoptosis induction (increased TUNEL staining and p53 expression, and decreased Bcl-2 expression). According to far-UV circular dichroism (CD) and molecular dynamic simulation data, VGB1 can adopt a helical structure. These results, for the first time, demonstrate that α1 helix region of VEGF-B recognizes both VEGFR-1 and VEGFR-2.
Collapse
|
8
|
Ehtesham S, Sariri R, Eidi A, Hosseinkhani S. Effect of Disulfide Bond Incorporation on the Structure and Activity of Endostatin Peptide. BIOCHEMISTRY (MOSCOW) 2018; 83:1388-1398. [PMID: 30482150 DOI: 10.1134/s0006297918110093] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
The structure and function of a 27-a.a. fragment of the N-terminal sequence of human endostatin (ES-Zn) were compared to those of the mutant peptide (ES-SSZn) obtained by adding Cys-Pro-Ala to the endostatin N-terminus and substituting Asn16 for Cys ensuring formation of a disulfide bond. Structural comparison of ES-Zn and ES-SSZn by far-UV circular dichroism (CD), intrinsic fluorescence, and molecular dynamics simulation methods revealed significant structural perturbations in ES-SSZn, such as elimination of the β-sheet conformer, modification of the N-terminal loop structure, and reorganization of dynamic properties of the entire peptide backbone. ES-SSZn was approximately 2 and 3 times less efficient than ES-Zn and the full-length human endostatin, respectively, in the induction of caspase-3-dependent apoptosis in human umbilical vein endothelial cells (HUVECs) in vitro (p < 0.05). In contrast, treatment of metastatic 4T1 breast tumors in mice with ES-Zn and ES-SSZn (5 mg/kg body weight daily) for 14 days resulted in similar regression of tumor size, comparable downregulation of angiogenesis (CD31 and CD34) and cell proliferation (Ki67), and therefore, the same extent of apoptosis induction (TUNEL, p53, and Bcl-2) for both peptides (as compared to the untreated controls). Western blot analysis of HUVEC and 4T1 tumor lysates revealed the same levels of suppression of key signaling mediators Akt and ERK1/2 by ES-Zn and ES-SSZn. Contrary to the earlier studies, our results showed that the function of the 1-27 endostatin fragment is independent of its overall structure. Stabilization of the N-terminal loop structure by the disulfide bond incorporation causes relief from structural deviations.
Collapse
Affiliation(s)
- S Ehtesham
- Department of Biology, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - R Sariri
- Department of Biology, Faculty of Sciences, University of Guilan, Rasht, Iran.
| | - A Eidi
- Department of Biology, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - S Hosseinkhani
- Department of Biochemistry, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran.
| |
Collapse
|
9
|
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]
|