Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 7.966
Filter
1.
Int J Biol Sci ; 20(7): 2454-2475, 2024.
Article in English | MEDLINE | ID: mdl-38725854

ABSTRACT

The emergence of Poly (ADP-ribose) polymerase inhibitors (PARPi) has marked the beginning of a precise targeted therapy era for ovarian cancer. However, an increasing number of patients are experiencing primary or acquired resistance to PARPi, severely limiting its clinical application. Deciphering the underlying mechanisms of PARPi resistance and discovering new therapeutic targets is an urgent and critical issue to address. In this study, we observed a close correlation between glycolysis, tumor angiogenesis, and PARPi resistance in ovarian cancer. Furthermore, we discovered that the natural compound Paris saponin VII (PS VII) partially reversed PARPi resistance in ovarian cancer and demonstrated synergistic therapeutic effects when combined with PARPi. Additionally, we found that PS VII potentially hindered glycolysis and angiogenesis in PARPi-resistant ovarian cancer cells by binding and stabilizing the expression of RORα, thus further inhibiting ECM1 and interfering with the VEGFR2/FAK/AKT/GSK3ß signaling pathway. Our research provides new targeted treatment for clinical ovarian cancer therapy and brings new hope to patients with PARPi-resistant ovarian cancer, effectively expanding the application of PARPi in clinical treatment.


Subject(s)
Diosgenin/analogs & derivatives , Glycolysis , Neovascularization, Pathologic , Ovarian Neoplasms , Saponins , Signal Transduction , Vascular Endothelial Growth Factor Receptor-2 , Female , Humans , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/metabolism , Vascular Endothelial Growth Factor Receptor-2/metabolism , Saponins/pharmacology , Saponins/therapeutic use , Signal Transduction/drug effects , Glycolysis/drug effects , Cell Line, Tumor , Neovascularization, Pathologic/drug therapy , Neovascularization, Pathologic/metabolism , Drug Resistance, Neoplasm/drug effects , Poly(ADP-ribose) Polymerase Inhibitors/pharmacology , Poly(ADP-ribose) Polymerase Inhibitors/therapeutic use , Animals , Mice, Nude , Mice , Angiogenesis
2.
Drug Dev Res ; 85(3): e22195, 2024 May.
Article in English | MEDLINE | ID: mdl-38704831

ABSTRACT

We investigated the angiogenesis-modulating ability of noscapine in vitro using osteosarcoma cell line (MG-63) and in vivo using a zebrafish model. MTT assay and the scratch wound healing assay were performed on the osteosarcoma cell line (MG-63) to analyze the cytotoxic effect and antimigrative ability of noscapine, respectively. We also observed the antiangiogenic ability of noscapine on zebrafish embryos by analyzing the blood vessels namely the dorsal aorta, and intersegmental vessels development at 24, 48, and 72 h postfertilization. Real-time polymerase chain reaction was used to analyze the hypoxia signaling molecules' gene expression in MG-63 cells and zebrafish embryos. The findings from the scratch wound healing demonstrated that noscapine stopped MG-63 cancer cells from migrating under both hypoxia and normoxia. Blood vessel development and the heart rate in zebrafish embryos were significantly reduced by noscapine under both hypoxia and normoxia which showed the hemodynamics impact of noscapine. Noscapine also downregulated the cobalt chloride (CoCl2) induced hypoxic signaling molecules' gene expression in MG-63 cells and zebrafish embryos. Therefore, noscapine may prevent MG-63 cancer cells from proliferating and migrating, as well as decrease the formation of new vessels and the production of growth factors linked to angiogenesis in vivo under both normoxic and hypoxic conditions.


Subject(s)
Hemodynamics , Neovascularization, Pathologic , Noscapine , Zebrafish , Animals , Humans , Noscapine/pharmacology , Cell Line, Tumor , Hemodynamics/drug effects , Neovascularization, Pathologic/drug therapy , Angiogenesis Inhibitors/pharmacology , Hypoxia , Cell Movement/drug effects , Embryo, Nonmammalian/drug effects , Osteosarcoma/drug therapy , Angiogenesis
3.
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
4.
Molecules ; 29(9)2024 Apr 29.
Article in English | MEDLINE | ID: mdl-38731540

ABSTRACT

Deferoxamine, an iron chelator used to treat diseases caused by excess iron, has had a Food and Drug Administration-approved status for many years. A large number of studies have confirmed that deferoxamine can reduce inflammatory response and promote angiogenesis. Blood vessels play a crucial role in sustaining vital life by facilitating the delivery of immune cells, oxygen, and nutrients, as well as eliminating waste products generated during cellular metabolism. Dysfunction in blood vessels may contribute significantly to the development of life-threatening diseases. Anti-angiogenesis therapy and pro-angiogenesis/angiogenesis strategies have been frequently recommended for various diseases. Herein, we describe the mechanism by which deferoxamine promotes angiogenesis and summarize its application in chronic wounds, bone repair, and diseases of the respiratory system. Furthermore, we discuss the drug delivery system of deferoxamine for treating various diseases, providing constructive ideas and inspiration for the development of new treatment strategies.


Subject(s)
Deferoxamine , Neovascularization, Physiologic , Deferoxamine/pharmacology , Deferoxamine/therapeutic use , Humans , Animals , Neovascularization, Physiologic/drug effects , Regeneration/drug effects , Wound Healing/drug effects , Neovascularization, Pathologic/drug therapy , Neovascularization, Pathologic/metabolism , Angiogenesis
5.
Biomacromolecules ; 25(5): 3087-3097, 2024 May 13.
Article in English | MEDLINE | ID: mdl-38584438

ABSTRACT

Heparan sulfate proteoglycans (HSPGs) play a crucial role in regulating cancer growth and migration by mediating interactions with growth factors. In this study, we developed a self-assembling peptide (S1) containing a sulfate group to simulate the contiguous sulfated regions (S-domains) in heparan sulfate for growth factor binding, aiming to sequester growth factors like VEGF. Spectral and structural studies as well as simulation studies suggested that S1 self-assembled into nanostructures similar to the heparan sulfate chains and effectively bound to VEGF. On cancer cell surfaces, S1 self-assemblies sequestered VEGF, leading to a reduction in VEGF levels in the medium, consequently inhibiting cancer cell growth, invasion, and angiogenesis. This study highlights the potential of self-assembling peptides to emulate extracellular matrix functions, offering insights for future cancer therapeutic strategies.


Subject(s)
Neoplasm Invasiveness , Peptides , Vascular Endothelial Growth Factor A , Humans , Vascular Endothelial Growth Factor A/metabolism , Peptides/chemistry , Peptides/pharmacology , Cell Movement/drug effects , Cell Line, Tumor , Heparitin Sulfate/chemistry , Neoplasms/drug therapy , Neoplasms/pathology , Neoplasms/metabolism , Cell Proliferation/drug effects , Neovascularization, Pathologic/drug therapy
6.
Int J Biol Macromol ; 267(Pt 1): 131409, 2024 May.
Article in English | MEDLINE | ID: mdl-38582478

ABSTRACT

Vessel normalization has proved imperative in tumor growth inhibition. In this work, biopolymer-based hybrid nanospheres capable of normalizing blood vessels were designed to improve the therapeutic effect of chemotherapeutic drugs. Zn0.4Fe2.6O4 nanoparticles (ZFO NPs) were synthesized, and were encapsulated in cross-inked chitosan (CS) along with a nitric oxide (NO) precursor, DETA NONOate, forming hybrid ZFO/NO@CS nanospheres highly stable in physiological environment. The structure, morphology and size of the nanospheres were characterized. The ZFO/NO@CS nanospheres could release NO under acidic conditions typical of intratumoral and intracellular environment. The results of related factors expression, wound healing and tube formation assays demonstrated that both the encapsulated ZFO NPs and the released NO were able to inhibit angiogenesis in tumors. The ZFO/NO@CS nanospheres enhanced the antitumor efficacy of the chemotherapeutic drug DOX by normalizing tumor vessels, as evidenced by in vivo experiments for CT26 tumor-bearing mice. By analyzing the contents of Fe in the tumor and different organs, the nanospheres were found to accumulate primarily at the tumor site. The blood analysis showed little side effect of the nanospheres. The ZFO/NO@CS nanospheres have great potential in improving tumor therapeutic effect when used in combination with chemotherapeutic drugs.


Subject(s)
Chitosan , Nanospheres , Chitosan/chemistry , Animals , Nanospheres/chemistry , Mice , Cell Line, Tumor , Nitric Oxide/metabolism , Doxorubicin/pharmacology , Doxorubicin/chemistry , Doxorubicin/administration & dosage , Neovascularization, Pathologic/drug therapy , Humans , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Drug Carriers/chemistry , Neoplasms/drug therapy , Neoplasms/blood supply , Neoplasms/pathology
7.
Int J Mol Sci ; 25(8)2024 Apr 16.
Article in English | MEDLINE | ID: mdl-38673959

ABSTRACT

Ovarian cancer poses a significant threat to patients in its advanced stages, often with limited treatment options available. In such cases, palliative management becomes the primary approach to maintaining a reasonable quality of life. Therefore, the administration of any medication that can benefit patients without a curative option holds potential. Resveratrol, a natural compound known for its in vitro anticancer activities, has generated contrasting results in vivo and human studies. In this study, we aimed to assess the anticancer effects of resveratrol on ovarian cancer cells grown on the chorioallantoic membrane (CAM) of chicken embryos. Two ovarian cancer cell lines, OVCAR-8 and SKOV-3, were cultured in collagen scaffolds for four days before being implanted on the CAM of chicken embryos on day 7. Different doses of resveratrol were applied to the CAM every two days for six days. Subsequently, CAM tissues were excised, fixed, and subjected to histological analysis. Some CAM tumours were extracted to analyse proteins through Western blotting. Our findings indicate that specific doses of resveratrol significantly reduce angiogenic activities, pNF-κB levels, and SLUG protein levels by using immunohistochemistry. These results suggest that resveratrol may have the potential to impact the behaviour of ovarian cancer CAM tumours, thereby warranting further consideration as a complementary treatment option for women with incurable ovarian cancer.


Subject(s)
Chorioallantoic Membrane , Ovarian Neoplasms , Resveratrol , Resveratrol/pharmacology , Chorioallantoic Membrane/drug effects , Animals , Female , Chick Embryo , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/pathology , Ovarian Neoplasms/metabolism , Humans , Cell Line, Tumor , Snail Family Transcription Factors/metabolism , Neovascularization, Pathologic/drug therapy , NF-kappa B/metabolism , Antineoplastic Agents, Phytogenic/pharmacology
8.
Int J Mol Sci ; 25(8)2024 Apr 17.
Article in English | MEDLINE | ID: mdl-38674018

ABSTRACT

Long-lasting anti-vascular endothelial growth factor (anti-VEGF) agents have become an option to reduce treatment frequency, with ongoing research exploring optimal responses and safety profiles. This review delves into molecular targets, pharmacological aspects, and strategies for achieving effective and enduring disease control in neovascular age-related macular degeneration (AMD). The molecular pathways involved in macular neovascularization, including angiogenesis and arteriogenesis, are explored. VEGF, PlGF, Ang-1, and Ang-2 play crucial roles in regulating angiogenesis, influencing vessel growth, maturation, and stability. The complex interplay of these factors, along with growth factors like TGFß and bFGF, contributes to the pathogenesis of neovascular membranes. Current anti-VEGF therapies, including bevacizumab, ranibizumab, aflibercept, brolucizumab, and faricimab, are discussed with a focus on their pharmacokinetics and clinical applications. Strategies to achieve sustained disease control in AMD involve smaller molecules, increased drug dosages, and novel formulations. This narrative review provides a comprehensive overview of the molecular targets and pharmacological aspects of neovascular AMD treatment.


Subject(s)
Angiogenesis Inhibitors , Macular Degeneration , Molecular Targeted Therapy , Humans , Macular Degeneration/drug therapy , Macular Degeneration/metabolism , Angiogenesis Inhibitors/therapeutic use , Angiogenesis Inhibitors/pharmacology , Molecular Targeted Therapy/methods , Vascular Endothelial Growth Factor A/metabolism , Vascular Endothelial Growth Factor A/antagonists & inhibitors , Animals , Neovascularization, Pathologic/drug therapy , Neovascularization, Pathologic/metabolism
9.
Int J Biol Macromol ; 267(Pt 2): 131546, 2024 May.
Article in English | MEDLINE | ID: mdl-38614172

ABSTRACT

Chitosan-based nanoparticles inevitably adsorb numerous proteins in the bloodstream, forming a protein corona that significantly influences their functionality. This study employed a pre-coated protein corona using cyclic Arg-Gly-Asp peptide (cRGD)-modified bovine serum albumin (BcR) to confer tumor-targeting capabilities on siVEGF-loaded chitosan-based nanoparticles (CsR/siVEGF NPs) and actively manipulated the serum protein corona composition to enhance their anti-tumor angiogenesis. Consequently, BcR effectively binds to the nanoparticles' surface, generating nanocarriers of appropriate size and stability that enhance the inhibition of endothelial cell proliferation, migration, invasion, and tube formation, as well as suppress tumor proliferation and angiogenesis in tumor-bearing nude mice. Proteomic analysis indicated a significant enrichment of serotransferrin, albumin, and proteasome subunit alpha type-1 in the protein corona of BcR-precoated NPs formed in the serum of tumor-bearing nude mice. Additionally, there was a decrease in proteins associated with complement activation, immunoglobulins, blood coagulation, and acute-phase responses. This modification resulted in an enhanced impact on anti-tumor angiogenesis, along with a reduction in opsonization and inflammatory responses. Therefore, pre-coating of nanoparticles with a functionalized albumin corona to manipulate the composition of serum protein corona emerges as an innovative approach to improve the delivery effectiveness of chitosan-based carriers for siVEGF, targeting the inhibition of tumor angiogenesis.


Subject(s)
Chitosan , Nanoparticles , Neovascularization, Pathologic , Protein Corona , Serum Albumin, Bovine , Chitosan/chemistry , Animals , Nanoparticles/chemistry , Mice , Humans , Protein Corona/chemistry , Serum Albumin, Bovine/chemistry , Neovascularization, Pathologic/drug therapy , Mice, Nude , Human Umbilical Vein Endothelial Cells/drug effects , Peptides, Cyclic/chemistry , Peptides, Cyclic/pharmacology , Cell Proliferation/drug effects , Drug Carriers/chemistry , Cattle , Vascular Endothelial Growth Factor A/metabolism , Xenograft Model Antitumor Assays , Cell Movement/drug effects , Cell Line, Tumor , Angiogenesis Inhibitors/pharmacology , Angiogenesis Inhibitors/chemistry , Angiogenesis
10.
Cell Death Dis ; 15(4): 292, 2024 Apr 24.
Article in English | MEDLINE | ID: mdl-38658527

ABSTRACT

Although bevacizumab (BVZ), a representative drug for anti-angiogenesis therapy (AAT), is used as a first-line treatment for patients with glioblastoma (GBM), its efficacy is notably limited. Whereas several mechanisms have been proposed to explain the acquisition of AAT resistance, the specific underlying mechanisms have yet to be sufficiently ascertained. Here, we established that inhibitor of differentiation 1 (ID1)high/activin Ahigh glioblastoma cell confers resistance to BVZ. The bipotent effect of activin A during its active phase was demonstrated to reduce vasculature dependence in tumorigenesis. In response to a temporary exposure to activin A, this cytokine was found to induce endothelial-to-mesenchymal transition via the Smad3/Slug axis, whereas prolonged exposure led to endothelial apoptosis. ID1 tumors showing resistance to BVZ were established to be characterized by a hypovascular structure, hyperpermeability, and scattered hypoxic regions. Using a GBM mouse model, we demonstrated that AAT resistance can be overcome by administering therapy based on a combination of BVZ and SB431542, a Smad2/3 inhibitor, which contributed to enhancing survival. These findings offer valuable insights that could contribute to the development of new strategies for treating AAT-resistant GBM.


Subject(s)
Activins , Angiogenesis Inhibitors , Bevacizumab , Drug Resistance, Neoplasm , Glioblastoma , Inhibitor of Differentiation Protein 1 , Glioblastoma/drug therapy , Glioblastoma/pathology , Glioblastoma/metabolism , Glioblastoma/blood supply , Humans , Animals , Inhibitor of Differentiation Protein 1/metabolism , Inhibitor of Differentiation Protein 1/genetics , Mice , Angiogenesis Inhibitors/pharmacology , Angiogenesis Inhibitors/therapeutic use , Activins/metabolism , Drug Resistance, Neoplasm/drug effects , Cell Line, Tumor , Bevacizumab/pharmacology , Bevacizumab/therapeutic use , Neovascularization, Pathologic/drug therapy , Neovascularization, Pathologic/metabolism , Neovascularization, Pathologic/pathology , Mice, Nude , Apoptosis/drug effects
11.
Clin Transl Med ; 14(4): e1648, 2024 04.
Article in English | MEDLINE | ID: mdl-38602256

ABSTRACT

BACKGROUND: Understanding how to modulate the microenvironment of tumors that are resistant to immune checkpoint inhibitors represents a major challenge in oncology.Here we investigate the ability of USP7 inhibitors to reprogram the tumor microenvironment (TME) by inhibiting secretion of vascular endothelial growth factor (VEGF) from fibroblasts. METHODS: To understand the role played by USP7 in the TME, we systematically evaluated the effects of potent, selective USP7 inhibitors on co-cultures comprising components of the TME, using human primary cells. We also evaluated the effects of USP7 inhibition on tumor growth inhibition in syngeneic models when dosed in combination with immune checkpoint inhibitors (ICIs). RESULTS: Abrogation of VEGF secretion from fibroblasts in response to USP7 inhibition resulted in inhibition of tumor neoangiogenesis and increased tumor recruitment of CD8-positive T-lymphocytes, leading to significantly improved sensitivity to immune checkpoint inhibitors. In syngeneic models, treatment with USP7 inhibitors led to striking tumor responses resulting in significantly improved survival. CONCLUSIONS: USP7-mediated reprograming of the TME is not linked to its previously characterized role in modulating MDM2 but does require p53 and UHRF1 in addition to the well-characterized VEGF transcription factor, HIF-1α. This represents a function of USP7 that is unique to fibroblasts, and which is not observed in cancer cells or other components of the TME. Given the potential for USP7 inhibitors to transform "immune desert" tumors into "immune responsive" tumors, this paves the way for a novel therapeutic strategy combining USP7 inhibitors with immune checkpoint inhibitors (ICIs).


Subject(s)
Neoplasms , Ubiquitin-Specific Peptidase 7 , Vascular Endothelial Growth Factor A , Humans , CCAAT-Enhancer-Binding Proteins/pharmacology , Fibroblasts/metabolism , Immune Checkpoint Inhibitors/pharmacology , Immune Checkpoint Inhibitors/therapeutic use , Neovascularization, Pathologic/drug therapy , Tumor Microenvironment , Ubiquitin-Specific Peptidase 7/antagonists & inhibitors
12.
PeerJ ; 12: e17222, 2024.
Article in English | MEDLINE | ID: mdl-38650654

ABSTRACT

Targeting tumor angiogenesis is an important approach in advanced tumor therapy. Here we investigated the effect of the suppressor of variegation 3-9 homolog 1 (SUV39H1) on tumor angiogenesis in oral squamous cell carcinoma (OSCC). The GEPIA database was used to analyze the expression of SUV39H1 in various cancer tissues. The expression of SUV39H1 in OSCC was detected by immunohistochemistry, and the correlation between SUV39H1 and Notch1 and microvascular density (MVD) was analyzed. The effect of SUV39H1 inhibition on OSCC was investigated in vivo by chaetocin treatment. The migration and tube formation of vascular endothelial cells by conditioned culture-medium of different treatments of oral squamous cell cells were measured. The transcriptional level of SUV39H1 is elevated in various cancer tissues. The transcription level of SUV39H1 in head and neck squamous cell carcinoma was significantly higher than that in control. Immunohistochemistry result showed increased SUV39H1 expression in OSCC, which was significantly correlated with T staging. The expression of SUV39H1 was significantly correlated with Notch1 and CD31. In vivo experiment chaetocin treatment significantly inhibit the growth of tumor, and reduce SUV39H1, Notch1, CD31 expression. The decreased expression of SUV39H1 in OSCC cells lead to the decreased expression of Notch1 and VEGF proteins, as well as the decreased migration and tube formation ability of vascular endothelial cells. Inhibition of Notch1 further enhance this effect. Our results suggest inhibition of SUV39H1 may affect angiogenesis by regulating Notch1 expression. This study provides a foundation for SUV39H1 as a potential therapeutic target for OSCC.


Subject(s)
Carcinoma, Squamous Cell , Methyltransferases , Mouth Neoplasms , Neovascularization, Pathologic , Receptor, Notch1 , Repressor Proteins , Humans , Receptor, Notch1/metabolism , Receptor, Notch1/genetics , Mouth Neoplasms/pathology , Mouth Neoplasms/metabolism , Mouth Neoplasms/drug therapy , Mouth Neoplasms/blood supply , Neovascularization, Pathologic/metabolism , Neovascularization, Pathologic/drug therapy , Neovascularization, Pathologic/pathology , Animals , Repressor Proteins/metabolism , Repressor Proteins/genetics , Methyltransferases/metabolism , Methyltransferases/antagonists & inhibitors , Methyltransferases/genetics , Carcinoma, Squamous Cell/pathology , Carcinoma, Squamous Cell/drug therapy , Carcinoma, Squamous Cell/metabolism , Carcinoma, Squamous Cell/blood supply , Cell Line, Tumor , Mice , Cell Movement/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Angiogenesis
13.
Chem Biol Drug Des ; 103(5): e14533, 2024 May.
Article in English | MEDLINE | ID: mdl-38684373

ABSTRACT

Hirudin is one of the specific inhibitors of thrombin, which has been confirmed to have strong bioactivities, including inhibiting tumors. However, the function and mechanism of hirudin and protease-activated receptor 1 (PAR-1) in diffuse large B-cell lymphoma (DLBCL) have not been clear. Detecting the expression PAR-1 in DLBCL tissues and cells by RT-qPCR and IHC. Transfected sh-NC, sh-PAR-1, or pcDNA3.1-PAR-1 in DLBCL cells or processed DLBCL cells through added thrombin, Vorapaxar, Recombinant hirudin (RH), or Na2S2O4 and co-culture with EA.hy926. And built DLBCL mice observed tumor growth. Detecting the expression of related genes by RT-qPCR, Western blot, IHC, and immunofluorescence, measured the cellular hypoxia with Hypoxyprobe-1 Kit, and estimated the cell inflammatory factors, proliferation, migration, invasion, and apoptosis by ELISA, CCK-8, flow cytometry, wound-healing and Transwell. Co-immunoprecipitation and pull-down measurement were used to verify the relationship. PAR-1 was highly expressed in DLBCL tissues and cells, especially in SUDHL2. Na2S2O4 induced SUDHL2 hypoxia, and PAR-1 did not influence thrombin-activated hypoxia. PAR-1 could promote SUDHL2 proliferation, migration, and invasion, and it was unrelated to cellular hypoxia. PAR-1 promoted proliferation, migration, and angiogenesis of EA.hy926 or SUDHL2 through up-regulation vascular endothelial growth factor (VEGF). RH inhibited tumor growth, cell proliferation, and migration, promoted apoptosis of DLBCL, and inhibited angiogenesis by down-regulating PAR-1-VEGF. RH inhibits proliferation, migration, and angiogenesis of DLBCL cells by down-regulating PAR-1-VEGF.


Subject(s)
Apoptosis , Cell Proliferation , Hirudins , Lymphoma, Large B-Cell, Diffuse , Neovascularization, Pathologic , Receptor, PAR-1 , Recombinant Proteins , Vascular Endothelial Growth Factor A , Humans , Hirudins/pharmacology , Receptor, PAR-1/metabolism , Receptor, PAR-1/antagonists & inhibitors , Lymphoma, Large B-Cell, Diffuse/metabolism , Lymphoma, Large B-Cell, Diffuse/drug therapy , Lymphoma, Large B-Cell, Diffuse/pathology , Animals , Vascular Endothelial Growth Factor A/metabolism , Vascular Endothelial Growth Factor A/genetics , Mice , Cell Line, Tumor , Neovascularization, Pathologic/drug therapy , Neovascularization, Pathologic/metabolism , Apoptosis/drug effects , Recombinant Proteins/pharmacology , Recombinant Proteins/metabolism , Cell Proliferation/drug effects , Cell Movement/drug effects , Angiogenesis
14.
J Nanobiotechnology ; 22(1): 209, 2024 Apr 25.
Article in English | MEDLINE | ID: mdl-38664830

ABSTRACT

BACKGROUND: Vasculogenic mimicry (VM), when microvascular channels are formed by cancer cells independent of endothelial cells, often occurs in deep hypoxic areas of tumors and contributes to the aggressiveness and metastasis of triple-negative breast cancer (TNBC) cells. However, well-developed VM inhibitors exhibit inadequate efficacy due to their low drug utilization rate and limited deep penetration. Thus, a cost-effective VM inhibition strategy needs to be designed for TNBC treatment. RESULTS: Herein, we designed a low-intensity focused ultrasound (LIFU) and matrix metalloproteinase-2 (MMP-2) dual-responsive nanoplatform termed PFP@PDM-PEG for the cost-effective and efficient utilization of the drug disulfiram (DSF) as a VM inhibitor. The PFP@PDM-PEG nanodroplets effectively penetrated tumors and exhibited substantial accumulation facilitated by PEG deshielding in a LIFU-mediated and MMP-2-sensitive manner. Furthermore, upon exposure to LIFU irradiation, DSF was released controllably under ultrasound imaging guidance. This secure and controllable dual-response DSF delivery platform reduced VM formation by inhibiting COL1/pro-MMP-2 activity, thereby significantly inhibiting tumor progression and metastasis. CONCLUSIONS: Considering the safety of the raw materials, controlled treatment process, and reliable repurposing of DSF, this dual-responsive nanoplatform represents a novel and effective VM-based therapeutic strategy for TNBC in clinical settings.


Subject(s)
Disulfiram , Lung Neoplasms , Matrix Metalloproteinase 2 , Nanoparticles , Neovascularization, Pathologic , Triple Negative Breast Neoplasms , Disulfiram/pharmacology , Triple Negative Breast Neoplasms/drug therapy , Triple Negative Breast Neoplasms/pathology , Matrix Metalloproteinase 2/metabolism , Animals , Female , Humans , Mice , Cell Line, Tumor , Lung Neoplasms/drug therapy , Lung Neoplasms/pathology , Lung Neoplasms/secondary , Nanoparticles/chemistry , Neovascularization, Pathologic/drug therapy , Mice, Inbred BALB C , Mice, Nude , Drug Repositioning , Ultrasonic Waves , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Antineoplastic Agents/therapeutic use
15.
Nat Commun ; 15(1): 3599, 2024 Apr 27.
Article in English | MEDLINE | ID: mdl-38678014

ABSTRACT

Targeting the supportive tumor microenvironment (TME) is an approach of high interest in cancer drug development. However, assessing TME-targeted drug candidates presents a unique set of challenges. We develop a comprehensive screening platform that allows monitoring, quantifying, and ranking drug-induced effects in self-organizing, vascularized tumor spheroids (VTSs). The confrontation of four human-derived cell populations makes it possible to recreate and study complex changes in TME composition and cell-cell interaction. The platform is modular and adaptable for tumor entity or genetic manipulation. Treatment effects are recorded by light sheet fluorescence microscopy and translated by an advanced image analysis routine in processable multi-parametric datasets. The system proved to be robust, with strong interassay reliability. We demonstrate the platform's utility for evaluating TME-targeted antifibrotic and antiangiogenic drugs side-by-side. The platform's output enabled the differential evaluation of even closely related drug candidates according to projected therapeutic needs.


Subject(s)
Breast Neoplasms , Microscopy, Fluorescence , Spheroids, Cellular , Tumor Microenvironment , Humans , Tumor Microenvironment/drug effects , Spheroids, Cellular/drug effects , Spheroids, Cellular/pathology , Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , Microscopy, Fluorescence/methods , Female , Cell Line, Tumor , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Drug Screening Assays, Antitumor/methods , Angiogenesis Inhibitors/pharmacology , Angiogenesis Inhibitors/therapeutic use , Neovascularization, Pathologic/drug therapy , Neovascularization, Pathologic/pathology
16.
Expert Opin Pharmacother ; 25(3): 255-262, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38591252

ABSTRACT

INTRODUCTION: Hepatocellular carcinoma (HCC) is a primary liver cancer that commonly arises in the background of chronic liver inflammation and/or cirrhosis. Chronic liver inflammation results in the production of different growth factors, remodeling of the microenvironment architecture into fibrosis, and eventually carcinogenesis. Overexpression of some growth factors has been associated with worse prognosis in patients with HCC. Targeted therapies against growth factors may disrupt cell signaling and the mechanisms that allow for cell survival (e.g. angiogenesis, proliferation, metastases). AREAS COVERED: We herein review potential growth factor targets of HCC and the limited research that exists regarding targeted therapy of these ligands and their receptors. We performed an extensive literature search to investigate preclinical studies, clinical research, and clinical trials. EXPERT OPINION: Systemic therapy for patients with HCC is continuing to evolve. Anti-angiogenic therapy holds the most promise among targeted therapy for growth factors among patients with HCC. Improving our understanding of growth factors in HCC will hopefully lead to the development of new targeted therapies and strategies for combination therapies with immune checkpoint inhibitors.


Subject(s)
Carcinoma, Hepatocellular , Intercellular Signaling Peptides and Proteins , Liver Neoplasms , Molecular Targeted Therapy , Humans , Carcinoma, Hepatocellular/drug therapy , Carcinoma, Hepatocellular/pathology , Carcinoma, Hepatocellular/metabolism , Liver Neoplasms/drug therapy , Liver Neoplasms/pathology , Liver Neoplasms/metabolism , Animals , Intercellular Signaling Peptides and Proteins/metabolism , Angiogenesis Inhibitors/therapeutic use , Angiogenesis Inhibitors/pharmacology , Antineoplastic Agents/therapeutic use , Antineoplastic Agents/pharmacology , Neovascularization, Pathologic/drug therapy , Prognosis , Drug Development
17.
Int J Biol Macromol ; 267(Pt 2): 131320, 2024 May.
Article in English | MEDLINE | ID: mdl-38569989

ABSTRACT

Macrofungi, a class of unique natural resources, are gaining popularity owing to their potential therapeutic benefits and edibility. From Fomitopsis officinalis, a medicinal macrofungus with anticancer activity, a homogeneous heteropolysaccharide (FOBP50-1) with a molecular weight of 2.21 × 104 g/mol has been extracted and purified. FOBP50-1 was found to be composed of 3-O-methylfucose, fucose, mannose, glucose, and galactose with a ratio of 1: 6.5: 4.4: 8.1: 18.2. The sugar fragments and structure of FOBP50-1 were investigated, which included →6)-α-d-Galp-(1→, →2,6)-α-d-Galp-(1→, →3)-α-l-Fucp-(1→, α-d-Glcp-(1→, →3)-ß-d-Manp-(1→, →6)-ß-d-Manp-(1→, 3-O-Me-α-l-Fucp-(1→, according to the UV, FT-IR, GC-MS, and NMR data. Besides the structure elucidation, FOBP50-1 showed promising antitumor activity in the zebrafish assays. The following mechanism examination discovered that FOBP50-1 interacted with TLR-4, PD-1, and VEGF to activate immunity and inhibit angiogenesis according to a series of cell, transgenic zebrafish, and surface plasmon resonance (SPR) experiments. The KD values indicating the association of FOBP50-1 with TLR-4, PD-1, and VEGF, were 4.69 × 10-5, 7.98 × 10-6, 3.04 × 10-6 M, respectively, in the SPR experiments. All investigations have demonstrated that the homogenous fungal polysaccharide FOBP50-1 has the potential to be turned into a tumor immunotherapy agent.


Subject(s)
Angiogenesis Inhibitors , Antineoplastic Agents , Fungal Polysaccharides , Zebrafish , Fungal Polysaccharides/chemistry , Fungal Polysaccharides/pharmacology , Fungal Polysaccharides/isolation & purification , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Antineoplastic Agents/isolation & purification , Angiogenesis Inhibitors/pharmacology , Angiogenesis Inhibitors/chemistry , Angiogenesis Inhibitors/isolation & purification , Humans , Coriolaceae/chemistry , Neovascularization, Pathologic/drug therapy , Vascular Endothelial Growth Factor A/metabolism , Mice , Angiogenesis
18.
ACS Appl Mater Interfaces ; 16(17): 21709-21721, 2024 May 01.
Article in English | MEDLINE | ID: mdl-38651381

ABSTRACT

Antiangiogenic therapy is an effective way to disrupt nutrient supply and starve tumors, but it is restricted by poor efficacy and negative feedback-induced tumor relapse. In this study, a neuropilin-1 (NRP-1)-targeted nanomedicine (designated as FPPT@Axi) is reported for spatiotemporal tumor suppression by combining photodynamic therapy (PDT) with antiangiogenesis. In brief, FPPT@Axi is prepared by utilizing an NRP-1-targeting chimeric peptide (Fmoc-K(PpIX)-PEG8-TKPRR) to encapsulate the antiangiogenic drug Axitinib (Axi). Importantly, the NRP-1-mediated targeting property enables FPPT@Axi to selectively concentrate at vascular endothelial and breast cancer cells, facilitating the production of reactive oxygen species (ROS) in situ for specific vascular disruption and enhanced cell apoptosis under light stimulation. Moreover, the codelivered Axi can further inhibit vascular endothelial growth factor receptor (VEGFR) to impair the negative feedback of PDT-induced tumor neovascularization. Consequently, FPPT@Axi spatiotemporally restrains the tumor growth through blocking angiogenesis, destroying tumor vessels, and inducing tumor apoptosis. Such an NRP-1-mediated targeting codelivery system sheds light on constructing an appealing candidate with translational potential by using clinically approved PDT and chemotherapy.


Subject(s)
Angiogenesis Inhibitors , Neovascularization, Pathologic , Neuropilin-1 , Photochemotherapy , Neuropilin-1/metabolism , Humans , Animals , Mice , Angiogenesis Inhibitors/pharmacology , Angiogenesis Inhibitors/chemistry , Neovascularization, Pathologic/drug therapy , Neovascularization, Pathologic/pathology , Female , Axitinib/pharmacology , Axitinib/chemistry , Axitinib/therapeutic use , Nanomedicine , Apoptosis/drug effects , Human Umbilical Vein Endothelial Cells , Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , Breast Neoplasms/metabolism , Mice, Inbred BALB C , Cell Line, Tumor , Photosensitizing Agents/pharmacology , Photosensitizing Agents/chemistry , Photosensitizing Agents/therapeutic use , Reactive Oxygen Species/metabolism , Mice, Nude
19.
Brain Tumor Pathol ; 41(2): 61-72, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38619734

ABSTRACT

Glioblastoma multiforme (GBM) acquires resistance to bevacizumab (Bev) treatment. Bev affects angiogenic factors other than vascular endothelial growth factor (VEGF), which are poorly understood. We investigated changes in angiogenic factors under and after Bev therapy, including angiopoietin-1 (ANGPT1), angiopoietin-2 (ANGPT2), placental growth factor (PLGF), fibroblast growth factor 2, and ephrin A2 (EphA2). Fifty-four GBM tissues, including 28 specimens from 14 cases as paired specimens from the same patient obtained in three settings: initial tumor resection (naïve Bev), tumors resected following Bev therapy (effective Bev), and recurrent tumors after Bev therapy (refractory Bev). Immunohistochemistry assessed their expressions in tumor vessels and its correlation with recurrent MRI patterns. PLGF expression was higher in the effective Bev group than in the naïve Bev group (p = 0.024) and remained high in the refractory Bev group. ANGPT2 and EphA2 expressions were higher in the refractory Bev group than in the naïve Bev group (p = 0.047 and 0.028, respectively). PLGF expression was higher in the refractory Bev group compared with the naïve Bev group for paired specimens (p = 0.036). PLGF was more abundant in T2 diffuse/circumscribe patterns (p = 0.046). This is the first study to evaluate angiogenic factors other than VEGF during effective and refractory Bev therapy in patient-derived specimens.


Subject(s)
Angiogenesis Inhibitors , Angiopoietin-2 , Bevacizumab , Brain Neoplasms , Glioblastoma , Neovascularization, Pathologic , Humans , Glioblastoma/drug therapy , Glioblastoma/pathology , Glioblastoma/surgery , Bevacizumab/therapeutic use , Brain Neoplasms/drug therapy , Brain Neoplasms/pathology , Male , Female , Middle Aged , Aged , Neovascularization, Pathologic/drug therapy , Adult , Angiopoietin-2/metabolism , Angiogenesis Inhibitors/therapeutic use , Placenta Growth Factor/metabolism , Antineoplastic Agents, Immunological/therapeutic use , Angiopoietin-1/metabolism , Neoplasm Recurrence, Local
20.
J Cancer Res Clin Oncol ; 150(5): 221, 2024 Apr 30.
Article in English | MEDLINE | ID: mdl-38687357

ABSTRACT

Vascular endothelial growth factor A (VEGF-A), a highly conserved dimeric glycoprotein, is a key regulatory gene and a marker molecule of angiogenesis. The upregulation of VEGF-A facilitates the process of tumor vascularization, thereby fostering the initiation and progression of malignant neoplasms. Many genes can adjust the angiogenesis of tumors by changing the expression of VEGF-A. In addition, VEGF-A also exhibits immune regulatory properties, which directly or indirectly suppresses the antitumor activity of immune cells. The emergence of VEGF-A-targeted therapy alone or in rational combinations has revolutionized the treatment of various cancers. This review discusses how diverse mechanisms in various tumors regulate VEGF-A expression to promote tumor angiogenesis and the role of VEGF-A in tumor immune microenvironment. The application of drugs targeting VEGF-A in tumor therapy is also summarized including antibody molecule drugs and traditional Chinese medicine.


Subject(s)
Molecular Targeted Therapy , Neoplasms , Neovascularization, Pathologic , Tumor Microenvironment , Vascular Endothelial Growth Factor A , Humans , Neoplasms/drug therapy , Neoplasms/metabolism , Neoplasms/pathology , Vascular Endothelial Growth Factor A/metabolism , Vascular Endothelial Growth Factor A/antagonists & inhibitors , Molecular Targeted Therapy/methods , Neovascularization, Pathologic/drug therapy , Neovascularization, Pathologic/metabolism , Animals , Gene Expression Regulation, Neoplastic , Angiogenesis Inhibitors/therapeutic use
SELECTION OF CITATIONS
SEARCH DETAIL
...