Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 3.003
Filter
1.
Anal Chem ; 96(22): 9007-9015, 2024 Jun 04.
Article in English | MEDLINE | ID: mdl-38778775

ABSTRACT

This study explores the synthesis and characterization of aggregation-induced emission enhancement (AIEE)-active gold nanoclusters (AuNCs), focusing on their near-infrared luminescence properties and potential applications in biological imaging. These AIEE-active AuNCs were synthesized via the NaBH4-mediated reduction of HAuCl4 in the presence of peptides. We systematically investigated the influence of the peptide sequence on the optical features of the AuNCs, highlighting the role of glutamic acid in enhancing their quantum yield (QY). Among the synthesized peptide-stabilized AuNCs, EECEE-stabilized AuNCs exhibited the maximum QY and a pronounced AIEE effect at pH 5.0, making them suitable for the luminescence imaging of intracellular lysosomes. The AIEE characteristic of the EECEE-stabilized AuNCs was demonstrated through examinations using transmission electron microscopy, dynamic light scattering, zeta potential analysis, and single-particle imaging. The formation of the EECEE-stabilized AuNCs was confirmed by size-exclusion chromatography and mass spectrometry. Spectroscopic and electrochemical examinations uncover the formation process of EECEE-stabilized AuNCs, comprising EECEE-mediated reduction, NaBH4-induced nucleation, complex aggregation, and subsequent cluster growth. Furthermore, we demonstrated the utility of these AuNCs as luminescent probes for intracellular lysosomal imaging, leveraging their pH-responsive AIEE behavior. Additionally, cyclic arginylglycylaspartic acid (RGD)-modified AIEE dots, derived from cyclic RGD-linked peptide-induced aggregation of EECEE-stabilized AuNCs, were developed for single- and two-photon luminescence imaging of αvß3 integrin receptor-positive cancer cells.


Subject(s)
Gold , Integrin alphaVbeta3 , Lysosomes , Metal Nanoparticles , Gold/chemistry , Lysosomes/chemistry , Lysosomes/metabolism , Integrin alphaVbeta3/metabolism , Integrin alphaVbeta3/analysis , Humans , Metal Nanoparticles/chemistry , Peptides/chemistry , Peptides/chemical synthesis , Photons , Optical Imaging
2.
Cell Commun Signal ; 22(1): 295, 2024 May 27.
Article in English | MEDLINE | ID: mdl-38802814

ABSTRACT

BACKGROUND: Colorectal cancer (CRC) commonly exhibits tolerance to cisplatin treatment, but the underlying mechanisms remain unclear. Within the tumor microenvironment, macrophages play a role in resisting the cytotoxic effects of chemotherapy by engaging in efferocytosis to clear apoptotic cells induced by chemotherapeutic agents. The involvement of extracellular vesicles (EVs), an intercellular communicator within the tumor microenvironment, in regulating the efferocytosis for the promotion of drug resistance has not been thoroughly investigated. METHODS: We constructed GFP fluorescent-expressing CRC cell lines (including GFP-CT26 and GFP-MC38) to detect macrophage efferocytosis through flow cytometric analysis. We isolated and purified CRC-secreted EVs using a multi-step ultracentrifugation method and identified them through electron microscopy and nanoflow cytometry. Proteomic analysis was conducted to identify the protein molecules carried by CRC-EVs. MFGE8 knockout CRC cell lines were constructed using CRISPR-Cas9, and their effects were validated through in vitro and in vivo experiments using Western blotting, immunofluorescence, and flow cytometric analysis, confirming that these EVs activate the macrophage αvß3-Src-FAK-STAT3 signaling pathway, thereby promoting efferocytosis. RESULTS: In this study, we found that CRC-derived EVs (CRC-EVs) enhanced macrophage efferocytosis of cisplatin-induced apoptotic CRC cells. Analysis of The Cancer Genome Atlas (TCGA) database revealed a high expression of the efferocytosis-associated gene MFGE8 in CRC patients, suggesting a poorer prognosis. Additionally, mass spectrometry-based proteomic analysis identified a high abundance of MFGE8 protein in CRC-EVs. Utilizing CRISPR-Cas9 gene edition system, we generated MFGE8-knockout CRC cells, demonstrating that their EVs fail to upregulate macrophage efferocytosis in vitro and in vivo. Furthermore, we demonstrated that MFGE8 in CRC-EVs stimulated macrophage efferocytosis by increasing the expression of αvß3 on the cell surface, thereby activating the intracellular Src-FAK-STAT3 signaling pathway. CONCLUSIONS: Therefore, this study highlighted a mechanism in CRC-EVs carrying MFGE8 activated the macrophage efferocytosis. This activation promoted the clearance of cisplatin-induced apoptotic CRC cells, contributing to CRC resistance against cisplatin. These findings provide novel insights into the potential synergistic application of chemotherapy drugs, EVs inhibitors, and efferocytosis antagonists for CRC treatment.


Subject(s)
Colorectal Neoplasms , Extracellular Vesicles , Macrophages , Phagocytosis , Extracellular Vesicles/metabolism , Extracellular Vesicles/genetics , Colorectal Neoplasms/pathology , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/genetics , Macrophages/metabolism , Humans , Animals , Cell Line, Tumor , Mice , STAT3 Transcription Factor/metabolism , STAT3 Transcription Factor/genetics , Signal Transduction , Cisplatin/pharmacology , Integrin alphaVbeta3/metabolism , Integrin alphaVbeta3/genetics , Efferocytosis
3.
Nanoscale ; 16(20): 9953-9965, 2024 May 23.
Article in English | MEDLINE | ID: mdl-38693876

ABSTRACT

Sonodynamic therapy (SDT) is an advanced non-invasive cancer treatment strategy with moderate tissue penetration, less invasiveness and a reliable curative effect. However, due to the low stability, potential bio-toxicity and lack of tumor targeting capability of most sonosensitizers, the vast clinical application of SDT has been challenging and limited. Therefore, it is desirable to develop a novel approach to implement sonosensitizers to SDT for cancer treatments. In this study, an amphiphilic polypeptide was designed to effectively encapsulate rose bengal (RB) as a model sonosensitizer to form peptido-nanomicelles (REPNs). The as-fabricated REPNs demonstrated satisfactory tumor targeting and fluorescence performances, which made them superb imaging tracers in vivo. In the meantime, they generated considerable amounts of reactive oxygen species (ROS) to promote tumor cell apoptosis under ultrasound irradiation and showed excellent anti-tumor performance without obvious side effects. These engineered nanomicelles in combination with medical ultrasound may be used to achieve integrin αvß3-targeted sonodynamic therapy against breast cancer, and it is also a promising non-invasive cancer treatment strategy for clinical translations.


Subject(s)
Breast Neoplasms , Integrin alphaVbeta3 , Micelles , Peptides , Reactive Oxygen Species , Ultrasonic Therapy , Integrin alphaVbeta3/metabolism , Female , Peptides/chemistry , Peptides/pharmacology , Breast Neoplasms/therapy , Breast Neoplasms/pathology , Breast Neoplasms/metabolism , Breast Neoplasms/drug therapy , Humans , Animals , Mice , Cell Line, Tumor , Reactive Oxygen Species/metabolism , Apoptosis/drug effects , Rose Bengal/chemistry , Rose Bengal/pharmacology , Mice, Inbred BALB C , Mice, Nude , Nanoparticles/chemistry , Nanoparticles/therapeutic use
4.
Biomed Pharmacother ; 175: 116714, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38761419

ABSTRACT

Cancer is one of the top 10 fatal diseases worldwide, among which advanced metastatic carcinoma has the highest mortality rate. Sunitinib and immune checkpoint blockers are commonly used to treat metastatic renal carcinoma with limited efficacy. Therefore, there is an urgent need to develop novel targeted therapies for metastatic renal cancer. In this study, we designed an antibody fusion protein, 57103, that simultaneously targeted the cluster of differentiation 24 (CD24), interleukin 4 receptor (IL-4R), and integrin receptors αvß3 and α5ß1. In vitro assays showed that 57103 significantly suppressed the proliferation, migration, invasion, colony formation, and adhesion abilities of renal cancer cells, resulting in a comprehensive and significant antitumor effect. Furthermore, 57103 inhibited angiogenesis, promoted THP1-derived M0-type macrophage phagocytosis, and enhanced the antibody-dependent cellular cytotoxicity of peripheral blood mononuclear and NK92MI-CD16a cells. In vivo experiments revealed significant inhibition of tumor growth in ACHN cell xenograft nude mice and an MC38-hCD24 tumor-bearing mouse model. Immunohistochemical analysis showed that 57103 decreased the proliferation and induced the apoptosis of renal cancer cells, while inhibiting angiogenesis. The MC38-hPDL1 and MC38-hCD24-hPDL1 tumor-bearing mouse models further offer the possibility of combining 57103 with the PDL1 antagonist atezolizumab. In conclusion, 57103 is a potential candidate drug for the treatment of metastatic renal carcinoma or PDL1-overexpressing cancer.


Subject(s)
Cell Proliferation , Integrin alphaVbeta3 , Kidney Neoplasms , Mice, Nude , Tumor Microenvironment , Animals , Humans , Tumor Microenvironment/drug effects , Cell Line, Tumor , Kidney Neoplasms/drug therapy , Kidney Neoplasms/pathology , Integrin alphaVbeta3/metabolism , Integrin alphaVbeta3/antagonists & inhibitors , Mice , Cell Proliferation/drug effects , Xenograft Model Antitumor Assays , Recombinant Fusion Proteins/pharmacology , Carcinoma, Renal Cell/drug therapy , Carcinoma, Renal Cell/pathology , Apoptosis/drug effects , Mice, Inbred BALB C , Cell Movement/drug effects , Neovascularization, Pathologic/drug therapy , Neovascularization, Pathologic/pathology
5.
Mol Med ; 30(1): 57, 2024 May 02.
Article in English | MEDLINE | ID: mdl-38698308

ABSTRACT

BACKGROUND: Ossification of the posterior longitudinal ligament (OPLL), an emerging heterotopic ossification disease, causes spinal cord compression, resulting in motor and sensory dysfunction. The etiology of OPLL remains unclear but may involve integrin αVß3 regulating the process of osteogenesis and angiogenesis. In this study, we focused on the role of integrin αVß3 in OPLL and explored the underlying mechanism by which the c(RGDyk) peptide acts as a potent and selective integrin αVß3 inhibitor to inhibit osteogenesis and angiogenesis in OPLL. METHODS: OPLL or control ligament samples were collected in surgery. For OPLL samples, RNA-sequencing results revealed activation of the integrin family, particularly integrin αVß3. Integrin αVß3 expression was detected by qPCR, Western blotting, and immunohistochemical analysis. Fluorescence microscopy was used to observe the targeted inhibition of integrin αVß3 by the c(RGDyk) peptide on ligaments fibroblasts (LFs) derived from patients with OPLL and endothelial cells (ECs). The effect of c(RGDyk) peptide on the ossification of pathogenic LFs was detected using qPCR, Western blotting. Alkaline phosphatase staining or alizarin red staining were used to test the osteogenic capability. The effect of the c(RGDyk) peptide on angiogenesis was determined by EC migration and tube formation assays. The effects of the c(RGDyk) peptide on heterotopic bone formation were evaluated by micro-CT, histological, immunohistochemical, and immunofluorescence analysis in vivo. RESULTS: The results indicated that after being treated with c(RGDyk), the osteogenic differentiation of LFs was significantly decreased. Moreover, the c(RGDyk) peptide inhibited the migration of ECs and thus prevented the nutritional support required for osteogenesis. Furthermore, the c(RGDyk) peptide inhibited ectopic bone formation in mice. Mechanistic analysis revealed that c(RGDyk) peptide could inhibit osteogenesis and angiogenesis in OPLL by targeting integrin αVß3 and regulating the FAK/ERK pathway. CONCLUSIONS: Therefore, the integrin αVß3 appears to be an emerging therapeutic target for OPLL, and the c(RGDyk) peptide has dual inhibitory effects that may be valuable for the new therapeutic strategy of OPLL.


Subject(s)
Integrin alphaVbeta3 , Ossification of Posterior Longitudinal Ligament , Osteogenesis , Integrin alphaVbeta3/metabolism , Integrin alphaVbeta3/antagonists & inhibitors , Humans , Osteogenesis/drug effects , Animals , Mice , Ossification of Posterior Longitudinal Ligament/metabolism , Ossification of Posterior Longitudinal Ligament/drug therapy , Male , Female , Middle Aged , Neovascularization, Pathologic/drug therapy , Neovascularization, Pathologic/metabolism , Fibroblasts/metabolism , Fibroblasts/drug effects , Neovascularization, Physiologic/drug effects , Cell Movement/drug effects , Disease Models, Animal , Oligopeptides/pharmacology , Oligopeptides/chemistry , Angiogenesis
6.
Bioorg Med Chem ; 107: 117759, 2024 Jun 01.
Article in English | MEDLINE | ID: mdl-38795572

ABSTRACT

Small molecule drugs sourced from natural products are pivotal for novel therapeutic discoveries. However, their clinical deployment is often impeded by non-specific activity and severe adverse effects. This study focused on 3-fluoro-10-hydroxy-Evodiamine (F-OH-Evo), a potent derivative of Evodiamine, whose development is curtailed due to suboptimal tumor selectivity and heightened cytotoxicity. By harnessing the remarkable stability, specificity, and αvß3 integrin affinity of c(RGDFK), a novel prodrug by conjugating F-OH-Evo with cRGD was synthesized. This innovative prodrug substantially enhanced the tumor-specific targeting of F-OH-Evo and improved the anti-tumor activities. Among them, compound 3c demonstrated the best selective inhibitory activity toward U87 cancer cells in vitro. It selectively enterd U87 cells by binding to αvß3 integrin, releasing the parent molecule under the dual response of ROS and GSH to exert inhibitory activity on topo I. The results highlight the potential of cRGD-conjugated prodrugs in targeted cancer therapy. This approach signifies a significant advancement in developing safer and more effective chemotherapy drugs, emphasizing the role of prodrug strategies in overcoming the limitations of traditional cancer treatments.


Subject(s)
Antineoplastic Agents , Drug Screening Assays, Antitumor , Peptides, Cyclic , Prodrugs , Humans , Prodrugs/chemistry , Prodrugs/pharmacology , Prodrugs/chemical synthesis , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemical synthesis , Peptides, Cyclic/chemistry , Peptides, Cyclic/pharmacology , Peptides, Cyclic/chemical synthesis , Molecular Structure , Structure-Activity Relationship , Cell Proliferation/drug effects , Cell Line, Tumor , Dose-Response Relationship, Drug , Drug Delivery Systems , Integrin alphaVbeta3/metabolism , Integrin alphaVbeta3/antagonists & inhibitors
7.
Cancer Res ; 84(10): 1543-1545, 2024 May 15.
Article in English | MEDLINE | ID: mdl-38745495

ABSTRACT

Nutrient stress accompanies several stages of tumor progression, including metastasis formation. Metabolic reprogramming is a hallmark of cancer, and it has been associated with stress tolerance and anchorage-independent cell survival. Adaptive responses are required to support cancer cell survival under these conditions. In this issue of Cancer Research, Nam and colleagues showed that the extracellular matrix (ECM) receptor integrin ß3 was upregulated in lung cancer cells in response to nutrient starvation, resulting in increased cell survival that was independent from ECM binding. Delving into the molecular mechanisms responsible for this, the authors found that integrin ß3 promoted glutamine metabolism and oxidative phosphorylation (OXPHOS) by activating a Src/AMPK/PGC1α signaling pathway. Importantly, in vivo experiments confirmed that OXPHOS inhibition suppressed tumor initiation in an orthotopic model of lung cancer, while ß3 knockout completely abrogated tumor initiation. These observations indicate that targeting signaling pathways downstream of αvß3 could represent a promising therapeutic avenue to prevent lung cancer progression and metastasis. See related article by Nam et al., p. 1630.


Subject(s)
Integrin alphaVbeta3 , Lung Neoplasms , Humans , Integrin alphaVbeta3/metabolism , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Lung Neoplasms/genetics , Lung Neoplasms/secondary , Animals , Signal Transduction , Mice , Oxidative Phosphorylation , Stress, Physiological , Nutrients/metabolism
8.
J Immunol ; 212(11): 1680-1692, 2024 Jun 01.
Article in English | MEDLINE | ID: mdl-38607278

ABSTRACT

Plasmacytoid dendritic cells (pDCs) are strongly implicated as a major source of IFN-I in systemic lupus erythematosus (SLE), triggered through TLR-mediated recognition of nucleic acids released from dying cells. However, relatively little is known about how TLR signaling and IFN-I production are regulated in pDCs. In this article, we describe a role for integrin αvß3 in regulating TLR responses and IFN-I production by pDCs in mouse models. We show that αv and ß3-knockout pDCs produce more IFN-I and inflammatory cytokines than controls when stimulated through TLR7 and TLR9 in vitro and in vivo. Increased cytokine production was associated with delayed acidification of endosomes containing TLR ligands, reduced LC3 conjugation, and increased TLR signaling. This dysregulated TLR signaling results in activation of B cells and promotes germinal center (GC) B cell and plasma cell expansion. Furthermore, in a mouse model of TLR7-driven lupus-like disease, deletion of αvß3 from pDCs causes accelerated autoantibody production and pathology. We therefore identify a pDC-intrinsic role for αvß3 in regulating TLR signaling and preventing activation of autoreactive B cells. Because αvß3 serves as a receptor for apoptotic cells and cell debris, we hypothesize that this regulatory mechanism provides important contextual cues to pDCs and functions to limit responses to self-derived nucleic acids.


Subject(s)
Autoimmunity , Dendritic Cells , Integrin alphaVbeta3 , Lupus Erythematosus, Systemic , Mice, Knockout , Signal Transduction , Toll-Like Receptor 7 , Animals , Mice , Dendritic Cells/immunology , Integrin alphaVbeta3/immunology , Integrin alphaVbeta3/metabolism , Autoimmunity/immunology , Toll-Like Receptor 7/immunology , Toll-Like Receptor 7/metabolism , Toll-Like Receptor 7/genetics , Lupus Erythematosus, Systemic/immunology , Signal Transduction/immunology , Mice, Inbred C57BL , Cytokines/metabolism , Cytokines/immunology , Toll-Like Receptor 9/immunology , Toll-Like Receptor 9/metabolism , B-Lymphocytes/immunology , Autoantibodies/immunology , Membrane Glycoproteins/immunology , Membrane Glycoproteins/metabolism , Lymphocyte Activation/immunology , Disease Models, Animal
9.
Int J Nanomedicine ; 19: 3071-3086, 2024.
Article in English | MEDLINE | ID: mdl-38562611

ABSTRACT

Introduction: The high mortality rate of malignant ovarian cancer is attributed to the absence of effective early diagnosis methods. The LHRH receptor is specifically overexpressed in most ovarian cancers, and the integrin αvß3 receptor is also overexpressed on the surface of ovarian cancer cells. In this study, we designed LHRH analogues (LHRHa)/RGD co-modified paclitaxel liposomes (LHRHa-RGD-LP-PTX) to target LHRH receptor-positive ovarian cancers more effectively and enhance the anti-ovarian cancer effects. Methods: LHRHa-RGD-LP-PTX liposomes were prepared using the thin film hydration method. The morphology, physicochemical properties, cellular uptake, and cell viability were assessed. Additionally, the cellular uptake mechanism of the modified liposomes was investigated using various endocytic inhibitors. The inhibitory effect of the formulations on tumor spheroids was observed under a microscope. The co-localization with lysosomes was visualized using confocal laser scanning microscopy (CLSM), and the in vivo tumor-targeting ability of the formulations was assessed using the IVIS fluorescent imaging system. Finally, the in vivo anti-tumor efficacy of the formulations was evaluated in the armpits of BALB/c nude mice. Results: The results indicated that LHRHa-RGD-LP-PTX significantly enhanced cellular uptake in A2780 cells, increased cytotoxicity, and hand a more potent inhibitory effect on tumor spheroids of A2780 cells. It also showed enhanced co-localization with endosomes or lysosome in A2780 cells, improved tumor-targeting capability, and demonstrated an enhanced anti-tumor effect in LHRHR-positive ovarian cancers. Conclusion: The designed LHRHa-RGD-LP-PTX liposomes significantly enhanced the tumor-targeting ability and therapeutic efficacy for LHRH receptor-positive ovarian cancers.


Subject(s)
Ovarian Neoplasms , Animals , Mice , Humans , Female , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/pathology , Liposomes/chemistry , Receptors, LHRH , Integrin alphaVbeta3 , Cell Line, Tumor , Mice, Nude , Paclitaxel/therapeutic use , Oligopeptides/chemistry
10.
Cancer Res ; 84(10): 1630-1642, 2024 May 15.
Article in English | MEDLINE | ID: mdl-38588407

ABSTRACT

Cancer stem/tumor-initiating cells display stress tolerance and metabolic flexibility to survive in a harsh environment with limited nutrient and oxygen availability. The molecular mechanisms underlying this phenomenon could provide targets to prevent metabolic adaptation and halt cancer progression. Here, we showed in cultured cells and live human surgical biopsies of non-small cell lung cancer that nutrient stress drives the expression of the epithelial cancer stem cell marker integrin αvß3 via upregulation of the ß3 subunit, resulting in a metabolic reprogramming cascade that allows tumor cells to thrive despite a nutrient-limiting environment. Although nutrient deprivation is known to promote acute, yet transient, activation of the stress sensor AMP-activated protein kinase (AMPK), stress-induced αvß3 expression via Src activation unexpectedly led to secondary and sustained AMPK activation. This resulted in the nuclear localization of peroxisome proliferator-activated receptor-gamma coactivator 1α (PGC1α) and upregulation of glutamine metabolism, the tricarboxylic acid cycle, and oxidative phosphorylation. Pharmacological or genetic targeting of this axis prevented lung cancer cells from evading the effects of nutrient stress, thereby blocking tumor initiation in mice following orthotopic implantation of lung cancer cells. These findings reveal a molecular pathway driven by nutrient stress that results in cancer stem cell reprogramming to promote metabolic flexibility and tumor initiation. SIGNIFICANCE: Upregulation of integrin αvß3, a cancer stem cell marker, in response to nutrient stress activates sustained AMPK/PGC1α signaling that induces metabolic reprogramming in lung cancer cells to support their survival. See related commentary by Rainero, p. 1543.


Subject(s)
Carcinoma, Non-Small-Cell Lung , Integrin alphaVbeta3 , Lung Neoplasms , Up-Regulation , Humans , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Lung Neoplasms/genetics , Animals , Integrin alphaVbeta3/metabolism , Mice , Carcinoma, Non-Small-Cell Lung/metabolism , Carcinoma, Non-Small-Cell Lung/pathology , Carcinoma, Non-Small-Cell Lung/genetics , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/metabolism , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/genetics , AMP-Activated Protein Kinases/metabolism , Stress, Physiological , Nutrients/metabolism , Cell Line, Tumor , Gene Expression Regulation, Neoplastic
11.
Eur J Nucl Med Mol Imaging ; 51(7): 2023-2035, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38376806

ABSTRACT

Integrin receptor αvß3 and gastrin-releasing peptide receptor (GRPR) expression of tumors could be detected using PET imaging with radiolabeled Arg-Gly-Asp (RGD) and the antagonistic bombesin analog RM26, respectively. The purpose of this study was to investigate the dual receptor-targeting property of the heterodimer RGD-RM26-03 (denoted as LNC1015), demonstrate the tumor diagnostic value of [68Ga]Ga-LNC1015 in preclinical experiments, and evaluate its preliminary clinical feasibility. METHODS: LNC1015 was designed and synthesized by linking cyclic RGD and the RM26 peptide. Preclinical pharmacokinetics were detected in a PC3 xenograft model using microPET and biodistribution studies. The clinical feasibility of [68Ga]Ga-LNC1015 PET/CT was performed in patients with breast cancer, and the results were compared with those of 18F-fluorodeoxyglucose (FDG). RESULTS: [68Ga]Ga-LNC1015 had good stability in saline for at least 2 h, and favorable binding affinity and specificity were demonstrated in vitro and in vivo. The tumor uptake and retention of [68Ga]Ga-LNC1015 during PET imaging were improved compared with its monomeric counterparts [68Ga]Ga-RGD and [68Ga]Ga-RM26 at all the time points examined. In our initial clinical studies, the tumor uptake and tumor-to-background ratio (TBR) of primary and metastatic lesions in [68Ga]Ga-LNC1015 PET/CT were significantly higher than those in [18F]FDG PET/CT, resulting in high lesion detection rate and tumor delineation. CONCLUSION: The dual targeting radiotracer [68Ga]Ga-LNC1015 showed significantly improved tumor uptake and retention, as well as lower liver uptake than [68Ga]Ga-RGD and [68Ga]Ga-RM26 monomer. The first-in-human study showed high TBRs in patients, suggesting favorable pharmacokinetics and high clinical feasibility for PET/CT imaging of cancer.


Subject(s)
Gallium Radioisotopes , Integrin alphaVbeta3 , Oligopeptides , Receptors, Bombesin , Receptors, Bombesin/metabolism , Humans , Animals , Mice , Female , Integrin alphaVbeta3/metabolism , Oligopeptides/pharmacokinetics , Oligopeptides/chemistry , Tissue Distribution , Male , Positron Emission Tomography Computed Tomography/methods , Radiochemistry , Middle Aged , Cell Line, Tumor , Radioactive Tracers , Radiopharmaceuticals/pharmacokinetics , Radiopharmaceuticals/chemical synthesis , Radiopharmaceuticals/chemistry , Chemistry Techniques, Synthetic , Breast Neoplasms/diagnostic imaging , Breast Neoplasms/metabolism
12.
J Ovarian Res ; 17(1): 50, 2024 Feb 23.
Article in English | MEDLINE | ID: mdl-38395907

ABSTRACT

BACKGROUND: Individual patients with ovarian cancer show remarkably different prognosis. Present prognostic models for ovarian cancer mainly focus on clinico-pathological parameters, so quantifiable prognostic markers at molecular level are urgently needed. Platelets contribute to ovarian cancer progression, but have not been considered as biomarkers likely due to their instability. Here, we aimed to search for a stable prognostic marker from platelet-treated ovarian cancer cells, and explore its functions and mechanisms. METHODS: Microarrays analysis was done with platelet-treated SKOV-3 ovarian cancer cells. Relevant studies were searched in the Gene Expression Omnibus (GEO) database. The candidate genes were determined by differentially expressed genes (DEGs), Venn diagram drawing, protein-protein interaction (PPI) network, Cox proportional hazards model and Kaplan-Meier analysis. The expression of TGFBI in clinical samples was assessed by immunehistochemical staining (IHC), and the association of TGFBI levels with the clinic-pathological characteristics and prognosis in ovarian cancer patients was evaluated by univariate and multivariate analysis. The functions of TGFBI were predicted using data from TCGA, and validated by in vitro and in vivo experiments. The mechanism exploration was performed based on proteomic analysis, molecular docking and intervention study. RESULTS: TGFBI was significantly higher expressed in the platelet-treated ovarian cancer cells. An analysis of bioinformatics data revealed that increased expression of TGFBI led to significant decrease of overall survival (OS), progression-free survival (PFS) and post-progression survival (PPS) in ovarian cancer patients. Tissue microarray results showed that TGFBI was an independent factor for ovarian cancer, and TGFBI expression predict poor prognosis. Functionally, TGFBI affected the migration and invasion of ovarian cancer cells by regulation of epithelial mesenchymal transition (EMT) markers (CDH1 and CDH2) and extracellular matrix (ECM) degradation proteins (MMP-2). Mechanistically, TGFBI phosphorylated PI3K and Akt by combining integrin αvß3. CONCLUSIONS: We found out TGFBI as a novel prognostic indicator for ovarian cancer patients. TGFBI could promote metastasis in ovarian cancer by EMT induction and ECM remodeling, which might be associated with the activation of integrin αvß3-PI3K-Akt signaling pathway.


Subject(s)
Integrin alphaVbeta3 , Ovarian Neoplasms , Transforming Growth Factor beta , Female , Humans , Extracellular Matrix Proteins/metabolism , Molecular Docking Simulation , Ovarian Neoplasms/genetics , Ovarian Neoplasms/metabolism , Ovarian Neoplasms/pathology , Phosphatidylinositol 3-Kinases/metabolism , Prognosis , Proteomics , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction , Transforming Growth Factor beta/metabolism
13.
Biochim Biophys Acta Mol Cell Res ; 1871(4): 119701, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38417588

ABSTRACT

Recent findings suggest that uncarboxylated osteocalcin (GluOC) promotes glucose and lipid metabolism via its putative receptor GPRC6A; however, its direct effect on adipocytes remains elusive. In this study, we elucidated the effects of GluOC on adipocytes, with an emphasis on the role of cell adhesion molecules. We determined that GluOC promoted the expression of adipocyte adhesion molecule (ACAM) and its transcription factor Krüppel-like factor 4 and enhanced the cortical actin filament assembly, which ameliorated lipid droplet hypertrophy. Additionally, GluOC upregulated the expression of integrin αVß3 and activation of focal adhesion kinase (FAK) and prevented insulin receptor substrate 1 (IRS1) degradation by inhibiting the ubiquitin-proteasome system via the FAK-PLC-PKC axis, which activated IRS1-Akt-mediated glucose transporter 4 (GLUT4) transport. Furthermore, we showed that GluOC elevated the expression of the insulin-independent glucose transporters GLUT1 and GLUT8, which facilitated insulin stimulation-independent glucose transport. The GluOC-induced activation of integrin αVß3 signaling promoted microtubule assembly, which improved glucose and lipid metabolism via its involvement in intracellular vesicular transport. GluOC treatment also suppressed collagen type 1 formation, which might prevent adipose tissue fibrosis in obese individuals. Overall, our results imply that GluOC promotes glucose and lipid metabolism via ACAM, integrin αVß3, and GLUT1 and 8 expression, directly affecting adipocytes.


Subject(s)
Glucose , Lipid Metabolism , Humans , Glucose/metabolism , Osteocalcin/metabolism , Osteocalcin/pharmacology , Lipid Metabolism/genetics , Glucose Transporter Type 1/metabolism , Integrin alphaVbeta3 , Adipocytes/metabolism , Insulin/metabolism , Cell Adhesion Molecules/metabolism
14.
Cell Rep ; 43(2): 113799, 2024 Feb 27.
Article in English | MEDLINE | ID: mdl-38367239

ABSTRACT

Schlemm's canal (SC) functions to maintain proper intraocular pressure (IOP) by draining aqueous humor and has emerged as a promising therapeutic target for glaucoma, the second-leading cause of irreversible blindness worldwide. However, our current understanding of the mechanisms governing SC development and functionality remains limited. Here, we show that vitronectin (VTN) produced by limbal macrophages promotes SC formation and prevents intraocular hypertension by activating integrin αvß3 signaling. Genetic inactivation of this signaling system inhibited the phosphorylation of AKT and FOXO1 and reduced ß-catenin activity and FOXC2 expression, thereby causing impaired Prox1 expression and deteriorated SC morphogenesis. This ultimately led to increased IOP and glaucomatous optic neuropathy. Intriguingly, we found that aged SC displayed downregulated integrin ß3 in association with dampened Prox1 expression. Conversely, FOXO1 inhibition rejuvenated the aged SC by inducing Prox1 expression and SC regrowth, highlighting a possible strategy by targeting VTN/integrin αvß3 signaling to improve SC functionality.


Subject(s)
Glaucoma , Hypertension , Optic Nerve Diseases , Humans , Aged , Integrin alphaVbeta3 , Schlemm's Canal , Macrophages
15.
Cells ; 13(4)2024 Feb 07.
Article in English | MEDLINE | ID: mdl-38391921

ABSTRACT

FGF9 is a potent mitogen and survival factor, but FGF9 protein levels are generally low and restricted to a few adult organs. Aberrant expression of FGF9 usually results in cancer. However, the mechanism of FGF9 action has not been fully established. Previous studies showed that FGF1 and FGF2 directly bind to integrin αvß3, and this interaction is critical for signaling functions (FGF-integrin crosstalk). FGF1 and FGF2 mutants defective in integrin binding were defective in signaling, whereas the mutants still bound to FGFR suppressed angiogenesis and tumor growth, indicating that they act as antagonists. We hypothesize that FGF9 requires direct integrin binding for signaling. Here, we show that docking simulation of the interaction between FGF9 and αvß3 predicted that FGF9 binds to the classical ligand-binding site of αvß3. We show that FGF9 bound to integrin αvß3 and generated FGF9 mutants in the predicted integrin-binding interface. An FGF9 mutant (R108E) was defective in integrin binding, activating FRS2α and ERK1/2, inducing DNA synthesis, cancer cell migration, and invasion in vitro. R108E suppressed DNA synthesis and activation of FRS2α and ERK1/2 induced by WT FGF9 (dominant-negative effect). These findings indicate that FGF9 requires direct integrin binding for signaling and that R108E has potential as an antagonist to FGF9 signaling.


Subject(s)
Integrin alphaVbeta3 , Mitogens , Integrin alphaVbeta3/metabolism , Ligands , Fibroblast Growth Factor 1 , Fibroblast Growth Factor 2 , DNA
16.
Nanoscale ; 16(8): 4063-4081, 2024 Feb 22.
Article in English | MEDLINE | ID: mdl-38334981

ABSTRACT

Active targeting strategies have been proposed to enhance the selective uptake of nanoparticles (NPs) by diseased cells, and recent experimental findings have proven the effectiveness of this approach. However, no mechanistic studies have yet revealed the atomistic details of the interactions between ligand-activated NPs and integrins. As a case study, here we investigate, by means of advanced molecular dynamics simulations (MD) and machine learning methods (namely equilibrium MD, binding free energy calculations and training of self-organized maps), the interaction of a cyclic-RGD-conjugated PEGylated TiO2 NP (the nanodevice) with the extracellular segment of integrin αVß3 (the target), the latter experimentally well-known to be over-expressed in several solid tumors. Firstly, we proved that the cyclic-RGD ligand binding to the integrin pocket is established and kept stable even in the presence of the cumbersome realistic model of the nanodevice. In this respect, the unsupervised machine learning analysis allowed a detailed comparison of the ligand/integrin binding in the presence and in the absence of the nanodevice, which unveiled differences in the chemical features. Then, we discovered that unbound cyclic RGDs conjugated to the NP largely contribute to the interactions between the nanodevice and the integrin. Finally, by increasing the density of cyclic RGDs on the PEGylated TiO2 NP, we observed a proportional enhancement of the nanodevice/target binding. All these findings can be exploited to achieve an improved targeting selectivity and cellular uptake, and thus a more successful clinical outcome.


Subject(s)
Integrin alphaVbeta3 , Neoplasms , Humans , Integrin alphaVbeta3/metabolism , Molecular Dynamics Simulation , Ligands , Protein Binding , Oligopeptides/chemistry , Machine Learning , Polyethylene Glycols/chemistry
17.
Nucl Med Biol ; 128-129: 108880, 2024.
Article in English | MEDLINE | ID: mdl-38330637

ABSTRACT

Patients with HER2-positive and triple negative breast cancer (TNBC) are associated with increased risk to develop metastatic disease including reoccurring disease that is resistant to standard and targeted therapies. The αVß3 has been implicated in BC including metastatic disease. The aims of this study were to investigate the potential of αVß3-targeted peptides to deliver radioactive payloads to BC tumors expressing αVß3 on the tumor cells or limited to the tumors' neovascular. Additionally, we aimed to assess the pharmacokinetic profile of the targeted α-particle therapy (TAT) agent [225Ac]Ac-DOTA-cRGDfK dimer peptide and the in vivo generated decay daughters. The expression of αVß3 in a HER2-positive and a TNBC cell line were evaluated using western blot analysis. The pharmacokinetics of [111In]In-DOTA-cRGDfK dimer, a surrogate for the TAT-agent, was evaluated in subcutaneous mouse tumor models. The pharmacokinetic of the TAT-agent [225Ac]Ac-DOTA-cRGDfK dimer and its decay daughters were evaluated in healthy mice. Selective uptake of [111In]In-DOTA-cRGDfK dimer was shown in subcutaneous tumor models using αVß3-positive tumor cells as well as αVß3-negative tumor cells where the expression is limited to the neovasculature. Pharmacokinetic studies demonstrated rapid accumulation in the tumors with clearance from non-target organs. Dosimetric analysis of [225Ac]Ac-DOTA-cRGDfK dimer showed the highest radiation absorbed dose to the kidneys, which included the contributions from the free in vivo generated decay daughters. This study shows the potential of delivering radioactive payloads to BC tumors that have αVß3 expression on the tumor cells as well as limited expression to the neovascular of the tumor. Furthermore, this work determines the radiation absorbed doses to normal organs/tissues and identified key organs that act as suppliers and receivers of the actinium-225 free in vivo generated α-particle-emitting decay daughters.


Subject(s)
Triple Negative Breast Neoplasms , Mice , Humans , Animals , Oligopeptides/pharmacokinetics , Peptides , Integrin alphaVbeta3/metabolism
18.
J Hazard Mater ; 466: 133650, 2024 Mar 15.
Article in English | MEDLINE | ID: mdl-38309170

ABSTRACT

Organophosphate esters (OPEs) can cause adverse biological effects through binding to integrin αvß3. However, few studies have focused on the binding activity and mechanism of OPEs to integrin αvß3. Herein, a comprehensive investigation of the mechanisms by which OPEs bind to integrin αvß3 and determination of the binding affinity were conducted by in vitro and in silico approaches: competitive binding assay as well as pharmacophore, molecular docking and QSAR modeling. The results showed that all 18 OPEs exhibited binding activities to integrin αvß3; moreover, hydrogen bonds were identified as crucial intermolecular interactions. In addition, essential factors, including the -P = O structure of OPEs, key amino acid residues and suitable cavity volume of integrin αvß3, were identified to contribute to the formation of hydrogen bonds. Moreover, aryl-OPEs exhibited a lower binding activity with integrin αvß3 than halogenated- and alkyl-OPEs. Ultimately, the QSAR model constructed in this study was effectively used to predict the binding affinity of OPEs to integrin αvß3, and the results suggest that some OPEs might pose potential risks in aquatic environments. The results of this study comprehensively elucidated the binding mechanism of OPEs to integrin αvß3, and supported the environmental risk management of these emerging pollutants.


Subject(s)
Esters , Integrin alphaVbeta3 , Pharmacophore , Binding, Competitive , China , Environmental Monitoring , Esters/chemistry , Flame Retardants , Integrin alphaVbeta3/chemistry , Integrin alphaVbeta3/metabolism , Molecular Docking Simulation , Organophosphates , Quantitative Structure-Activity Relationship
19.
Int Dent J ; 74(3): 597-606, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38184457

ABSTRACT

OBJECTIVES: The aim of this study was to investigate the molecular mechanism underlying odontoblast damage repair in dentin hypersensitivity (DH) and the role of Yes-associated protein (YAP) in this process. METHODS: The DH model was constructed in Sprague-Dawley (SD) rats, and the in vivo expression of Piezo1, Integrin αvß3, YAP, and dentin sialophosphoprotein (DSPP) was detected by immunohistochemistry. COMSOL Multiphysics software was used to simulate the dentinal tubule fluid flow velocity and corresponding fluid shear stress (FSS) on the odontoblast processes. MDPC-23 cells were cultured in vitro and loaded with a peristaltic pump for 1 hour at FSS values of 0.1, 0.3, 0.5, and 0.7 dyne/cm2. The expression of Piezo1, Integrin αvß3, and YAP was detected by immunofluorescence. Verteporfin (a YAP-specific inhibitor) was utilised to confirm the effect of YAP on the expression of dentineogenesis-related protein under FSS. RESULTS: The level and duration of external mechanical stimuli have an effect on the functional expression of odontoblasts. In DH, the harder the food that is chewed, the faster the flow of the dentinal tubule fluid and the greater the FSS on the odontoblast processes. The expression of Piezo1, Integrin αvß3, and YAP can be promoted when the FSS is less than 0.3 dyne/cm2. After YAP inhibition, the DSPP protein expression level was reduced at 0.3 dyne/cm2 FSS. CONCLUSIONS: These results suggest that appropriate FSS can enhance the expression of odontoblast-related factors in odontoblasts via the Piezo1-Integrin αvß3-YAP mechanotransduction pathway and the YAP appears to play an essential role in the response of odontoblasts to external mechanical stimuli.


Subject(s)
Dentin Sensitivity , Disease Models, Animal , Odontoblasts , Rats, Sprague-Dawley , YAP-Signaling Proteins , Odontoblasts/metabolism , Animals , Rats , Phosphoproteins/metabolism , Integrin alphaVbeta3/metabolism , Stress, Mechanical , Extracellular Matrix Proteins/metabolism , Sialoglycoproteins/metabolism , Ion Channels/metabolism , Immunohistochemistry , Adaptor Proteins, Signal Transducing/metabolism , Verteporfin/pharmacology , Verteporfin/therapeutic use , Male , Membrane Proteins
20.
Eur J Nucl Med Mol Imaging ; 51(6): 1544-1557, 2024 May.
Article in English | MEDLINE | ID: mdl-38276986

ABSTRACT

PURPOSE: Several studies have demonstrated the advantages of heterodimers over their corresponding monomers due to the multivalency effect. This effect leads to an increased number of effective targeted receptors and, consequently, improved tumor uptake. Fibroblast activation protein (FAP) and integrin αvß3 are found to be overexpressed in different components of the tumor microenvironment. In our pursuit of enhancing tumor uptake and retention, we designed and developed a novel peptidic heterodimer that synergistically targets both FAP and integrin αvß3. METHODS: FAP-RGD was synthesized from FAP-2286 and c(RGDfK) through a multi-step organic synthesis. The dual receptor binding property of 68Ga-FAP-RGD was investigated by cell uptake and competitive binding assays. Preclinical pharmacokinetics were determined in HT1080-FAP and U87MG tumor models using micro-positron emission tomography/computed tomography (micro-PET/CT) and biodistribution studies. The antitumor efficacy of 177Lu-FAP-RGD was assessed in U87MG tumor models. The radiation exposure and clinical diagnostic performance of 68 Ga-FAP-RGD were evaluated in healthy volunteers and cancer patients. RESULTS: Bi-specific radiotracer 68Ga-FAP-RGD exhibited high binding affinity for both FAP and integrin αvß3. In comparison to 68Ga-FAP-2286 and 68Ga-RGDfK, 68Ga-FAP-RGD displayed enhanced tumor uptake and longer tumor retention time in preclinical models. 177Lu-FAP-RGD could efficiently suppress the growth of U87MG tumor in vivo when applied at an activity of 18.5 and 29.6 MBq. The effective dose of 68Ga-FAP-RGD was 1.06 × 10-2 mSv/MBq. 68Ga-FAP-RGD demonstrated low background activity and stable accumulation in most neoplastic lesions up to 3 h. CONCLUSION: Taking the advantages of multivalency effect, the bi-specific radiotracer 68Ga-FAP-RGD showed superior tumor uptake and retention compared to its corresponding monomers. Preclinical studies with 68Ga- or 177Lu-labeled FAP-RGD showed favorable image contrast and effective antitumor responses. Despite the excellent performance of 68Ga-FAP-RGD in clinical diagnosis, experimental efforts are currently underway to optimize the structure of FAP-RGD to increase its potential for clinical application in endoradiotherapy.


Subject(s)
Endopeptidases , Integrin alphaVbeta3 , Membrane Proteins , Positron Emission Tomography Computed Tomography , Serine Endopeptidases , Animals , Female , Humans , Mice , Cell Line, Tumor , Dimerization , Endopeptidases/metabolism , Endopeptidases/pharmacology , Gallium Radioisotopes/chemistry , Integrin alphaVbeta3/chemistry , Integrin alphaVbeta3/metabolism , Membrane Proteins/metabolism , Membrane Proteins/pharmacology , Oligopeptides/chemistry , Oligopeptides/pharmacokinetics , Positron Emission Tomography Computed Tomography/methods , Radioactive Tracers , Radiopharmaceuticals/pharmacokinetics , Radiopharmaceuticals/chemistry , Serine Endopeptidases/metabolism , Tissue Distribution , Peptides/metabolism , Peptides/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...