Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 11.497
Filter
1.
Anticancer Res ; 44(6): 2377-2392, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38821582

ABSTRACT

BACKGROUND/AIM: Oral squamous cell carcinoma (OSCC) is characterized by early metastasis, clinical resistance and poor prognosis. Recently, we showed that aggressive OSCC cells co-express endothelial cell markers and can form tube-like structures, known as vasculogenic mimicry (VM), a process associated with poor prognosis in head and neck cancers. Given the limited success of current antiangiogenic therapy in treating OSCC, this study sought to explore the efficiency of these drugs in targeting an ex vivo model of VM. MATERIALS AND METHODS: OSCC cell lines from the tongue and floor of the mouth in addition to human endothelial cells were used. The treatments comprised a set of clinically relevant antiangiogenic drugs: sorafenib, sunitinib, and axitinib, which were administered in different doses. Multiple ex vivo approaches including cell tubulogenesis, proliferation, apoptosis, and migration assays were used. RESULTS: Although these drugs inhibited the formation of endothelial cell capillaries, they showed clear differential effects on OSCC cell-derived VM and cell morphology. Sorafenib inhibited the tubulogenesis of aggressive OSCC cells compared with the limited effect of sunitinib and axitinib. Furthermore, our data consistently demonstrated a preferential efficacy of certain drugs over others. Sorafenib and sunitinib exhibited anti-cancer effects on tumor cell proliferation, apoptosis, and cell migration, compared with the limited effect of axitinib. CONCLUSION: The antiangiogenic drugs, except sorafenib, had limited effect on VM formation in vitro and exhibited varying anti-cancer effects on OSCC cells. These data support the notion that VM formation may in part explain the development of drug resistance in OSCC cells.


Subject(s)
Angiogenesis Inhibitors , Axitinib , Cell Movement , Cell Proliferation , Mouth Neoplasms , Neovascularization, Pathologic , Sorafenib , Sunitinib , Humans , Angiogenesis Inhibitors/pharmacology , Angiogenesis Inhibitors/therapeutic use , Mouth Neoplasms/drug therapy , Mouth Neoplasms/pathology , Mouth Neoplasms/blood supply , Mouth Neoplasms/metabolism , Cell Line, Tumor , Neovascularization, Pathologic/drug therapy , Neovascularization, Pathologic/pathology , Sorafenib/pharmacology , Sorafenib/therapeutic use , Sunitinib/pharmacology , Sunitinib/therapeutic use , Cell Proliferation/drug effects , Cell Movement/drug effects , Axitinib/pharmacology , Apoptosis/drug effects , Carcinoma, Squamous Cell/drug therapy , Carcinoma, Squamous Cell/pathology , Carcinoma, Squamous Cell/blood supply , Carcinoma, Squamous Cell/metabolism , Endothelial Cells/drug effects , Endothelial Cells/pathology , Endothelial Cells/metabolism , Phenylurea Compounds/pharmacology , Phenylurea Compounds/therapeutic use , Niacinamide/analogs & derivatives , Niacinamide/pharmacology , Niacinamide/therapeutic use
2.
J Histochem Cytochem ; 72(5): 329-352, 2024 May.
Article in English | MEDLINE | ID: mdl-38733294

ABSTRACT

Lactate and ATP formation by aerobic glycolysis, the Warburg effect, is considered a hallmark of cancer. During angiogenesis in non-cancerous tissue, proliferating stalk endothelial cells (ECs) also produce lactate and ATP by aerobic glycolysis. In fact, all proliferating cells, both non-cancer and cancer cells, need lactate for the biosynthesis of building blocks for cell growth and tissue expansion. Moreover, both non-proliferating cancer stem cells in tumors and leader tip ECs during angiogenesis rely on glycolysis for pyruvate production, which is used for ATP synthesis in mitochondria through oxidative phosphorylation (OXPHOS). Therefore, aerobic glycolysis is not a specific hallmark of cancer but rather a hallmark of proliferating cells and limits its utility in cancer therapy. However, local treatment of angiogenic eye conditions with inhibitors of glycolysis may be a safe therapeutic option that warrants experimental investigation. Most types of cells in the eye such as photoreceptors and pericytes use OXPHOS for ATP production, whereas proliferating angiogenic stalk ECs rely on glycolysis for lactate and ATP production. (J Histochem Cytochem XX.XXX-XXX, XXXX).


Subject(s)
Adenosine Triphosphate , Neoplasms , Neovascularization, Pathologic , Humans , Adenosine Triphosphate/metabolism , Adenosine Triphosphate/biosynthesis , Neoplasms/metabolism , Neoplasms/pathology , Neoplasms/blood supply , Neoplasms/drug therapy , Animals , Neovascularization, Pathologic/metabolism , Neovascularization, Pathologic/pathology , Glycolysis , Eye Diseases/metabolism , Eye Diseases/pathology , Oxidative Phosphorylation
3.
Bull Math Biol ; 86(7): 75, 2024 May 17.
Article in English | MEDLINE | ID: mdl-38758501

ABSTRACT

The landscape of computational modeling in cancer systems biology is diverse, offering a spectrum of models and frameworks, each with its own trade-offs and advantages. Ideally, models are meant to be useful in refining hypotheses, to sharpen experimental procedures and, in the longer run, even for applications in personalized medicine. One of the greatest challenges is to balance model realism and detail with experimental data to eventually produce useful data-driven models. We contribute to this quest by developing a transparent, highly parsimonious, first principle in silico model of a growing avascular tumor. We initially formulate the physiological considerations and the specific model within a stochastic cell-based framework. We next formulate a corresponding mean-field model using partial differential equations which is amenable to mathematical analysis. Despite a few notable differences between the two models, we are in this way able to successfully detail the impact of all parameters in the stability of the growth process and on the eventual tumor fate of the stochastic model. This facilitates the deduction of Bayesian priors for a given situation, but also provides important insights into the underlying mechanism of tumor growth and progression. Although the resulting model framework is relatively simple and transparent, it can still reproduce the full range of known emergent behavior. We identify a novel model instability arising from nutrient starvation and we also discuss additional insight concerning possible model additions and the effects of those. Thanks to the framework's flexibility, such additions can be readily included whenever the relevant data become available.


Subject(s)
Bayes Theorem , Computer Simulation , Mathematical Concepts , Models, Biological , Neoplasms , Stochastic Processes , Systems Biology , Humans , Neoplasms/pathology , Neovascularization, Pathologic/pathology
4.
Clin Exp Pharmacol Physiol ; 51(7): e13868, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38745265

ABSTRACT

Cervical cancer (CC) is a gynaecological malignancy tumour that seriously threatens women's health. Recent evidence has identified that interferon regulatory factor 5 (IRF5), a nucleoplasm shuttling protein, is a pivotal transcription factor regulating the growth and metastasis of various human tumours. This study aimed to investigate the function and molecular basis of IRF5 in CC development. IRF5, protein phosphatase 6 catalytic subunit (PPP6C) and methyltransferase-like 3 (METTL3) mRNA levels were evaluated by quantitative real-time (qRT)-polymerase chain reaction (PCR). IRF5, PPP6C, METTL3, B-cell lymphoma 2 and Bax protein levels were detected using western blot. Cell proliferation, migration, invasion, angiogenesis and apoptosis were determined by using colony formation, 5-ethynyl-2'-deoxyuridine (EdU), transwell, tube formation assay and flow cytometry assay, respectively. Glucose uptake and lactate production were measured using commercial kits. Xenograft tumour assay in vivo was used to explore the role of IRF5. After JASPAR predication, binding between IRF5 and PPP6C promoter was verified using chromatin immunoprecipitation and dual-luciferase reporter assays. Moreover, the interaction between METTL3 and IRF5 was verified using methylated RNA immunoprecipitation (MeRIP). IRF5, PPP6C and METTL3 were highly expressed in CC tissues and cells. IRF5 silencing significantly inhibited cell proliferation, migration, invasion, angiogenesis and glycolytic metabolism in CC cells, while induced cell apoptosis. Furthermore, the absence of IRF5 hindered tumour growth in vivo. At the molecular level, IRF5 might bind with PPP6C to positively regulate the expression of PPP6C mRNA. Meanwhile, IRF5 was identified as a downstream target of METTL3-mediated m6A modification. METTL3-mediated m6A modification of mRNA might promote CC malignant progression by regulating PPP6C, which might provide a promising therapeutic target for CC treatment.


Subject(s)
Cell Proliferation , Disease Progression , Interferon Regulatory Factors , Methyltransferases , Up-Regulation , Uterine Cervical Neoplasms , Humans , Female , Uterine Cervical Neoplasms/genetics , Uterine Cervical Neoplasms/pathology , Uterine Cervical Neoplasms/metabolism , Methyltransferases/genetics , Methyltransferases/metabolism , Interferon Regulatory Factors/genetics , Interferon Regulatory Factors/metabolism , Cell Line, Tumor , Animals , Cell Proliferation/genetics , Mice , Gene Expression Regulation, Neoplastic , Apoptosis/genetics , Cell Movement/genetics , Mice, Nude , Neoplasm Invasiveness , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/pathology , Neovascularization, Pathologic/metabolism
5.
Oncoimmunology ; 13(1): 2356942, 2024.
Article in English | MEDLINE | ID: mdl-38778816

ABSTRACT

Brain metastasis is the most devasting form of lung cancer. Recent studies highlight significant differences in the tumor microenvironment (TME) between lung cancer brain metastasis (LCBM) and primary lung cancer, which contribute significantly to tumor progression and drug resistance. Cancer-associated fibroblasts (CAFs) are the major component of pro-tumor TME with high plasticity. However, the lineage composition and function of CAFs in LCBM remain elusive. By reanalyzing single-cell RNA sequencing (scRNA-seq) data (GSE131907) from lung cancer patients with different stages of metastasis comprising primary lesions and brain metastasis, we found that CAFs undergo distinctive lineage transition during LCBM under a hypoxic situation, which is directly driven by hypoxia-induced HIF-2α activation. Transited CAFs enhance angiogenesis through VEGF pathways, trigger metabolic reprogramming, and promote the growth of tumor cells. Bulk RNA sequencing data was utilized as validation cohorts. Multiplex immunohistochemistry (mIHC) assay was performed on four paired samples of brain metastasis and their primary lung cancer counterparts to validate the findings. Our study revealed a novel mechanism of lung cancer brain metastasis featuring HIF-2α-induced lineage transition and functional alteration of CAFs, which offers potential therapeutic targets.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors , Brain Neoplasms , Cancer-Associated Fibroblasts , Lung Neoplasms , Tumor Microenvironment , Humans , Lung Neoplasms/pathology , Lung Neoplasms/secondary , Lung Neoplasms/metabolism , Lung Neoplasms/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Basic Helix-Loop-Helix Transcription Factors/genetics , Cancer-Associated Fibroblasts/metabolism , Cancer-Associated Fibroblasts/pathology , Brain Neoplasms/secondary , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Brain Neoplasms/genetics , Mice , Animals , Cell Line, Tumor , Phenotype , Cell Lineage , Gene Expression Regulation, Neoplastic , Neovascularization, Pathologic/pathology , Neovascularization, Pathologic/metabolism , Neovascularization, Pathologic/genetics , Single-Cell Analysis
6.
BMC Cancer ; 24(1): 633, 2024 May 23.
Article in English | MEDLINE | ID: mdl-38783271

ABSTRACT

BACKGROUND: PD-L1 overexpression is commonly observed in various malignancies and is strongly correlated with poor prognoses for cancer patients. Moreover, PD-L1 has been shown to play a significant role in promoting angiogenesis and epithelial-mesenchymal transition (EMT) processes across different cancer types. METHODS: The relationship between PD-L1 and vasculogenic mimicry as well as epithelial-mesenchymal transition (EMT) was explored by bioinformatics approach and immunohistochemistry. The functions of PD-L1 in regulating the expression of ZEB1 and the EMT process were assessed by Western blotting and q-PCR assays. The impact of PD-L1 on the migratory and proliferative capabilities of A549 and H1299 cells was evaluated through wound healing, cell invasion, and CCK8 assays following siRNA-mediated PD-L1 knockdown. Tube formation assay was utilized to evaluate the presence of VM structures. RESULTS: In this study, increased PD-L1 expression was observed in A549 and H1299 cells compared to normal lung epithelial cells. Immunohistochemical analysis revealed a higher prevalence of VM structures in the PD-L1-positive group compared to the PD-L1-negative group. Additionally, high PD-L1 expression was also found to be significantly associated with advanced TNM stage and increased metastasis. Following PD-L1 knockdown, NSCLC cells exhibited a notable reduction in their ability to form tube-like structures. Moreover, the levels of key EMT and VM-related markers, including N-cadherin, MMP9, VE-cadherin, and VEGFA, were significantly decreased, while E-cadherin expression was upregulated. In addition, the migration and proliferation capacities of both cell lines were significantly inhibited after PD-L1 or ZEB1 knockdown. CONCLUSIONS: Knockdown PD-L1 can inhibit ZEB1-mediated EMT, thereby hindering the formation of VM in NSCLC.


Subject(s)
B7-H1 Antigen , Carcinoma, Non-Small-Cell Lung , Cell Movement , Cell Proliferation , Epithelial-Mesenchymal Transition , Gene Knockdown Techniques , Lung Neoplasms , Neovascularization, Pathologic , Zinc Finger E-box-Binding Homeobox 1 , Humans , Epithelial-Mesenchymal Transition/genetics , Zinc Finger E-box-Binding Homeobox 1/genetics , Zinc Finger E-box-Binding Homeobox 1/metabolism , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/pathology , Carcinoma, Non-Small-Cell Lung/metabolism , B7-H1 Antigen/metabolism , B7-H1 Antigen/genetics , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Lung Neoplasms/metabolism , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/metabolism , Neovascularization, Pathologic/pathology , Cell Line, Tumor , Cell Proliferation/genetics , Cell Movement/genetics , Gene Expression Regulation, Neoplastic , Male , Female , A549 Cells , Middle Aged
7.
Cancer Lett ; 592: 216931, 2024 Jun 28.
Article in English | MEDLINE | ID: mdl-38701892

ABSTRACT

The intricate role of inducible nitric oxide synthase (iNOS) in cancer pathophysiology has garnered significant attention, highlighting the complex interplay between tumorigenesis, immune response, and cellular metabolism. As an enzyme responsible for producing nitric oxide (NO) in response to inflammatory stimuli. iNOS is implicated in various aspects of cancer development, including DNA damage, angiogenesis, and evasion of apoptosis. This review synthesizes the current findings from both preclinical and clinical studies on iNOS across different cancer types, reflecting the variability depending on cellular context and tumor microenvironment. We explore the molecular mechanisms by which iNOS modulates cancer cell growth, survival, and metastasis, emphasizing its impact on immune surveillance and response to treatment. Additionally, the potential of targeting iNOS as a therapeutic strategy in cancer treatment is examined. By integrating insights from recent advances, this review aims to elucidate the significant role of iNOS in cancer and pave the way for novel diagnostic and therapeutic approaches.


Subject(s)
Disease Progression , Neoplasms , Nitric Oxide Synthase Type II , Tumor Microenvironment , Humans , Nitric Oxide Synthase Type II/metabolism , Neoplasms/pathology , Neoplasms/enzymology , Animals , Nitric Oxide/metabolism , Neovascularization, Pathologic/pathology
8.
J Cancer Res Clin Oncol ; 150(5): 257, 2024 May 16.
Article in English | MEDLINE | ID: mdl-38753184

ABSTRACT

PURPOSE: Breast cancer metastasis relies on cellular invasion and angiogenesis facilitated by the downregulation of metastatic suppressor proteins like Cluster of Differentiation 82 (CD82). Currently, no medicines target multiple systems to prevent metastatic progression through CD82 upregulation. This study screened for plant extracts displaying effects on cell proliferation, invasion, and CD82 expression in breast cancer cells, and in vivo angiogenesis, and further correlated between the biological activities and effect on CD82 expression. METHODS: Seventeen ethanolic plant extracts were screened for their effect on cell proliferation (against MDA-MB-231 and MCF-7 breast cancer and Hek293 kidney cells), cell invasion and effect on CD82 expression in metastatic MDA-MB-231 cells. Selected extracts were further evaluated for in vivo anti-angiogenesis. RESULTS: Extracts displayed varying antiproliferative activity against the different cell lines, and those that showed selectivity indexes (SI) > 0.5 against MDA-MB-231 were selected for anti-invasion evaluation. Buddleja saligna Willd. (BS), Combretum apiculatum Sond. (CA), Foeniculum vulgare, Greyia radlkoferi, Gunnera perpensa and Persicaria senegalensis (Meisn.) Soják (PS) displayed 50% inhibitory concentration (IC50) values of 44.46 ± 3.46, 74.00 ± 4.48, 180.43 ± 4.51, 96.97 ± 2.29, 55.29 ± 9.88 and 243.60 ± 2.69 µg/mL, respectively against MDA-MB-231, and compared to Hek293 showed SI of 0.9, 0.7, 1.4, 1.1, 2.2 and 0.5. Significant invasion inhibition was observed at both 20 and 40 µg/mL for BS (94.10 ± 0.74 and 96.73 ± 0.95%) and CA (87.42 ± 6.54 and 98.24 ± 0.63%), whereas GR (14.91 ± 1.62 and 41 ± 1.78%) and PS (36.58 ± 0.54 and 51.51 ± 0.83%), only showed significant inhibition at 40 µg/mL, and FV (< 5% inhibition) and GP (10 ± 1.03 and 22 ± 1.31%) did not show significant inhibition at both concentrations. Due to the significant anti-invasive activity of BS, CA and PS at 40 µg/mL, these extracts were further evaluated for their potential to stimulate CD82. BS showed significant (p < 0.05) reduction in CD82 at 20 and 40 µg/mL (13.2 ± 2.2% and 20.3 ± 1.5% decrease, respectively), whereas both CA and PS at 20 µg/mL increased (p < 0.05) CD82 expression (16.4 ± 0.8% and 5.4 ± 0.6% increase, respectively), and at 40 µg/mL significantly reduced CD82 expression (23.4 ± 3.1% and 11.2 ± 2.9% decrease, respectively). Using the yolk sac membrane assay, BS (59.52 ± 4.12 and 56.72 ± 3.13% newly formed vessels) and CA (83.33 ± 3.17 and 74.00 ± 2.12%) at both 20 and 40 µg/egg showed significant (p < 0.001) angiogenesis inhibition, with BS showing statistical similar activity to the positive control, combretastatin A4 (10 nmol/egg), whereas PS only displayed significant (p < 0.001) angiogenesis stimulation at 40 µg/egg (120.81 ± 3.34% newly formed vessels). CONCLUSION: BS exhibits antiproliferative, anti-invasive, and anti-angiogenic activity despite inhibiting CD82, suggesting an alternative mode of action. CA at 20 µg/mL shows moderate anti-invasive and anti-angiogenic potential by stimulating CD82, while at 40 µg/mL it still displays these properties but inhibits CD82, suggesting an additional mode of action. PS, with the least antiproliferative activity, stimulates CD82 and inhibits angiogenesis at 20 µg/mL but inhibits CD82 and increases angiogenesis at 40 µg/mL, indicating CD82 targeting as a major mode of action. Future studies should explore breast cancer xenograft models to assess the extracts' impact on CD82 expression and angiogenesis in the tumor microenvironment, along with isolating bioactive compounds from the extracts.


Subject(s)
Breast Neoplasms , Cell Proliferation , Kangai-1 Protein , Neoplasm Invasiveness , Neovascularization, Pathologic , Plant Extracts , Humans , Breast Neoplasms/pathology , Breast Neoplasms/drug therapy , Breast Neoplasms/metabolism , Cell Proliferation/drug effects , Plant Extracts/pharmacology , Female , Animals , Neovascularization, Pathologic/drug therapy , Neovascularization, Pathologic/pathology , Neovascularization, Pathologic/prevention & control , Kangai-1 Protein/metabolism , Plants, Medicinal/chemistry , HEK293 Cells , Cell Line, Tumor , Ethanol/chemistry , Ethanol/pharmacology , Chick Embryo , Neoplasm Metastasis , Chorioallantoic Membrane/drug effects , Angiogenesis
9.
J Exp Clin Cancer Res ; 43(1): 135, 2024 May 04.
Article in English | MEDLINE | ID: mdl-38702792

ABSTRACT

BACKGROUND: Rhabdomyosarcoma (RMS) is a rare malignancy and the most common soft tissue sarcoma in children. Vasculogenic mimicry (VM) is a novel tumor microcirculation model different from traditional tumor angiogenesis, which does not rely on endothelial cells to provide sufficient blood supply for tumor growth. In recent years, VM has been confirmed to be closely associated with tumor progression. However, the ability of RMS to form VM has not yet been reported. METHODS: Immunohistochemistry, RT-qPCR and western blot were used to test the expression level of SNAI2 and its clinical significance. The biological function in regulating vasculogenic mimicry and malignant progression of SNAI2 was examined both in vitro and in vivo. Mass spectrometry, co-immunohistochemistry, immunofluorescence staining, and ubiquitin assays were performed to explore the regulatory mechanism of SNAI2. RESULTS: Our study indicated that SNAI2 was abnormally expressed in patients with RMS and RMS cell lines and promoted the proliferation and metastasis of RMS. Through cell tubule formation experiments, nude mice Matrigel plug experiments, and immunohistochemistry (IHC), we confirmed that RMS can form VM and that SNAI2 promotes the formation of VM. Due to SNAI2 is a transcription factor that is not easily drugged, we used Co-IP combined with mass spectrometry to screen for the SNAI2-binding protein USP7 and TRIM21. USP7 depletion inhibited RMS VM formation, proliferation and metastasis by promoting SNAI2 degradation. We further demonstrated that TRIM21 is expressed at low levels in human RMS tissues and inhibits VM in RMS cells. TRIM21 promotes SNAI2 protein degradation through ubiquitination in the RMS. The deubiquitinase USP7 and E3 ligase TRIM21 function in an antagonistic rather than competitive mode and play a key role in controlling the stability of SNAI2 to determine the VM formation and progression of RMS. CONCLUSION: Our findings reveal a previously unknown mechanism by which USP7 and TRIM21 balance the level of SNAI2 ubiquitination, determining RMS vasculogenic mimicry, proliferation, and migration. This new mechanism may provide new targeted therapies to inhibit the development of RMS by restoring TRIM21 expression or inhibiting USP7 expression in RMS patients with high SNAI2 protein levels.


Subject(s)
Neovascularization, Pathologic , Rhabdomyosarcoma , Ribonucleoproteins , Snail Family Transcription Factors , Ubiquitin-Specific Peptidase 7 , Humans , Snail Family Transcription Factors/metabolism , Snail Family Transcription Factors/genetics , Animals , Mice , Ubiquitin-Specific Peptidase 7/metabolism , Ubiquitin-Specific Peptidase 7/genetics , Rhabdomyosarcoma/metabolism , Rhabdomyosarcoma/pathology , Rhabdomyosarcoma/genetics , Neovascularization, Pathologic/metabolism , Neovascularization, Pathologic/pathology , Female , Disease Progression , Cell Proliferation , Male , Homeostasis , Cell Line, Tumor , Mice, Nude , Ubiquitin-Protein Ligases/metabolism , Ubiquitin-Protein Ligases/genetics , Ubiquitination
10.
Cancer Treat Rev ; 127: 102749, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38714074

ABSTRACT

Cancer cells merely exist in isolation; rather, they exist in an intricate microenvironment composed of blood vessels, signalling molecules, immune cells, stroma, fibroblasts, and the ECM. The TME provides a setting that is favourable for the successful growth and survivance of tumors. Angiogenesis is a multifaceted process that is essential for the growth, invasion, and metastasis of tumors. TME can be visualized as a "concert hall," where various cellular and non-cellular factors perform in a "symphony" to orchestrate tumor angiogenesis and create "Havoc" instead of "Harmony". In this review, we comprehensively summarized the involvement of TME in regulating tumor angiogenesis. Especially, we have focused on immune cells and their secreted factors, inflammatory cytokines and chemokines, and their role in altering the TME. We have also deciphered the crosstalk among various cell types that further aids the process of tumor angiogenesis. Additionally, we have highlighted the limitations of existing anti-angiogenic therapy and discussed various potential strategies that could be used to overcome these challenges and improve the efficacy of anti-angiogenic therapy.


Subject(s)
Neoplasms , Neovascularization, Pathologic , Tumor Microenvironment , Humans , Neovascularization, Pathologic/pathology , Neoplasms/pathology , Neoplasms/blood supply , Neoplasms/drug therapy , Animals , Angiogenesis Inhibitors/therapeutic use , Angiogenesis Inhibitors/pharmacology , Angiogenesis
11.
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
12.
Sci Rep ; 14(1): 10539, 2024 05 08.
Article in English | MEDLINE | ID: mdl-38719941

ABSTRACT

Abnormal angiogenesis leads to tumor progression and metastasis in colorectal cancer (CRC). This study aimed to elucidate the association between angiogenesis-related genes, including VEGF-A, ANGPT-1, and ANGPT-2 with both metastatic and microsatellite alterations at selected tetranucleotide repeats (EMAST) subtypes of CRC. We conducted a thorough assessment of the ANGPT-1, ANGPT-2, and VEGF-A gene expression utilizing publicly available RNA sequencing and microarray datasets. Then, the experimental validation was performed in 122 CRC patients, considering their disease metastasis and EMAST+/- profile by using reverse transcription polymerase chain reaction (RT-PCR). Subsequently, a competing endogenous RNA (ceRNA) network associated with these angiogenesis-related genes was constructed and analyzed. The expression level of VEGF-A and ANGPT-2 genes were significantly higher in tumor tissues as compared with normal adjacent tissues (P-value < 0.001). Nevertheless, ANGPT-1 had a significantly lower expression in tumor samples than in normal colon tissue (P-value < 0.01). We identified a significantly increased VEGF-A (P-value = 0.002) and decreased ANGPT-1 (P-value = 0.04) expression in EMAST+ colorectal tumors. Regarding metastasis, a significantly increased VEGF-A and ANGPT-2 expression (P-value = 0.001) and decreased ANGPT-1 expression (P-value < 0.05) were established in metastatic CRC patients. Remarkably, co-expression analysis also showed a strong correlation between ANGPT-2 and VEGF-A gene expressions. The ceRNA network was constructed by ANGPT-1, ANGPT-2, VEGF-A, and experimentally validated miRNAs (hsa-miR-190a-3p, hsa-miR-374c-5p, hsa-miR-452-5p, and hsa-miR-889-3p), lncRNAs (AFAP1-AS1, KCNQ1OT1 and MALAT1), and TFs (Sp1, E2F1, and STAT3). Network analysis revealed that colorectal cancer is amongst the 82 significant pathways. We demonstrated a significant differential expression of VEGF-A and ANGPT-1 in colorectal cancer patients exhibiting the EMAST+ phenotype. This finding provides novel insights into the molecular pathogenesis of colorectal cancer, specifically in EMAST subtypes. Yet, the generalization of in silico findings to EMAST+ colorectal cancer warrants future experimental investigations. In the end, this study proposes that the EMAST biomarker could serve as an additional perspective on CMS4 biology which is well-defined by activated angiogenesis and worse overall survival.


Subject(s)
Angiopoietin-1 , Angiopoietin-2 , Colorectal Neoplasms , Gene Expression Regulation, Neoplastic , Neovascularization, Pathologic , Vascular Endothelial Growth Factor A , Humans , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , Colorectal Neoplasms/metabolism , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/metabolism , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/pathology , Angiopoietin-1/genetics , Angiopoietin-1/metabolism , Angiopoietin-2/genetics , Angiopoietin-2/metabolism , Male , Female , Middle Aged , Neoplasm Metastasis , Aged , Microsatellite Repeats/genetics , Gene Expression Profiling , Gene Regulatory Networks , Angiogenesis
13.
Mol Cancer ; 23(1): 94, 2024 May 08.
Article in English | MEDLINE | ID: mdl-38720298

ABSTRACT

BACKGROUND: The hypoxic tumor microenvironment is a key factor that promotes metabolic reprogramming and vascular mimicry (VM) in ovarian cancer (OC) patients. ESM1, a secreted protein, plays an important role in promoting proliferation and angiogenesis in OC. However, the role of ESM1 in metabolic reprogramming and VM in the hypoxic microenvironment in OC patients has not been determined. METHODS: Liquid chromatography coupled with tandem MS was used to analyze CAOV3 and OV90 cells. Interactions between ESM1, PKM2, UBA2, and SUMO1 were detected by GST pull-down, Co-IP, and molecular docking. The effects of the ESM1-PKM2 axis on cell glucose metabolism were analyzed based on an ECAR experiment. The biological effects of the signaling axis on OC cells were detected by tubule formation, transwell assay, RT‒PCR, Western blot, immunofluorescence, and in vivo xenograft tumor experiments. RESULTS: Our findings demonstrated that hypoxia induces the upregulation of ESM1 expression through the transcription of HIF-1α. ESM1 serves as a crucial mediator of the interaction between PKM2 and UBA2, facilitating the SUMOylation of PKM2 and the subsequent formation of PKM2 dimers. This process promotes the Warburg effect and facilitates the nuclear translocation of PKM2, ultimately leading to the phosphorylation of STAT3. These molecular events contribute to the promotion of ovarian cancer glycolysis and vasculogenic mimicry. Furthermore, our study revealed that Shikonin effectively inhibits the molecular interaction between ESM1 and PKM2, consequently preventing the formation of PKM2 dimers and thereby inhibiting ovarian cancer glycolysis, fatty acid synthesis and vasculogenic mimicry. CONCLUSION: Our findings demonstrated that hypoxia increases ESM1 expression through the transcriptional regulation of HIF-1α to induce dimerization via PKM2 SUMOylation, which promotes the OC Warburg effect and VM.


Subject(s)
Carrier Proteins , Fatty Acids , Membrane Proteins , Neoplasm Proteins , Ovarian Neoplasms , Thyroid Hormone-Binding Proteins , Thyroid Hormones , Tumor Microenvironment , Female , Humans , Ovarian Neoplasms/metabolism , Ovarian Neoplasms/pathology , Ovarian Neoplasms/genetics , Animals , Thyroid Hormones/metabolism , Mice , Membrane Proteins/metabolism , Membrane Proteins/genetics , Cell Line, Tumor , Fatty Acids/metabolism , Neoplasm Proteins/metabolism , Neoplasm Proteins/genetics , Carrier Proteins/metabolism , Carrier Proteins/genetics , Warburg Effect, Oncologic , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Gene Expression Regulation, Neoplastic , Neovascularization, Pathologic/metabolism , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/pathology , Xenograft Model Antitumor Assays , Cell Proliferation , Proteoglycans
14.
Nanoscale ; 16(21): 10350-10365, 2024 May 30.
Article in English | MEDLINE | ID: mdl-38739006

ABSTRACT

Tumour cells secrete various proangiogenic factors like VEGF, PDGF, and EGF that result in the formation of highly vascularized tumours with an immunosuppressive tumour microenvironment. As tumour growth and metastasis are highly dependent on angiogenesis, targeting tumour vasculature along with rapidly dividing tumour cells is a potential approach for cancer treatment. Here, we specifically engineered sub-100 sized nanomicelles (DTX-CA4 NMs) targeting proliferation and angiogenesis using an esterase-sensitive phosphocholine-tethered docetaxel conjugate of lithocholic acid (LCA) (PC-LCA-DTX) and a poly(ethylene glycol) (PEG) derivative of an LCA-combretastatin A4 conjugate (PEG-LCA-CA4). DTX-CA4 NMs effectively inhibit the tumour growth in syngeneic (CT26) and xenograft (HCT116) colorectal cancer models, inhibit tumour recurrence, and enhance the percentage survival in comparison with individual drug-loaded NMs. DTX-CA4 NMs enhance the T cell-mediated anti-tumour immune response and DTX-CA4 NMs in combination with an immune checkpoint inhibitor, anti-PDL1 antibody, enhance the anti-tumour response. We additionally showed that DTX-CA4 NMs effectively attenuate the production of ceramide-1-phosphate, a key metabolite of the sphingolipid pathway, by downregulating the expression of ceramide kinase at both transcriptional and translational levels. Therefore, this study presents the engineering of effective DTX-CA4 NMs for targeting the tumour microenvironment that can be explored further for clinical applications.


Subject(s)
Cell Proliferation , Ceramides , Docetaxel , Micelles , Neovascularization, Pathologic , Animals , Ceramides/chemistry , Ceramides/pharmacology , Humans , Mice , Cell Proliferation/drug effects , Docetaxel/pharmacology , Docetaxel/chemistry , Neovascularization, Pathologic/drug therapy , Neovascularization, Pathologic/metabolism , Neovascularization, Pathologic/pathology , Lithocholic Acid/chemistry , Lithocholic Acid/pharmacology , Polyethylene Glycols/chemistry , Cell Line, Tumor , Mice, Inbred BALB C , Stilbenes/chemistry , Stilbenes/pharmacology , HCT116 Cells , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Antineoplastic Agents/chemical synthesis , Tumor Microenvironment/drug effects , Nanoparticles/chemistry , Xenograft Model Antitumor Assays , Female , Angiogenesis
15.
Biochim Biophys Acta Mol Basis Dis ; 1870(5): 167196, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38653358

ABSTRACT

RNA modification plays important roles in various physiological and pathological process. LAGE3 is a component of EKC/KEOPS complex, which is probably involved in the formation of a threonylcarbamoyl group on adenosine at position 37 (t(6)A37) in tRNAs, but its exact role in HCC is less studied. Our study reveals that LAGE3 exhibits upregulated expression in HCC compared with normal hepatocellular tissue. High expression of LAGE3 promotes hepatocellular cell proliferation and migration. Further investigations suggest that the increased expression of LAGE3 cloud lead to upregulated VEGFA secretion and angiogenesis in HCC. The mechanistic study reveals LAGE3 is required for the VEGFA mRNA stability. This research may open new avenues for diagnosis and targeted therapy in HCC.


Subject(s)
Carcinoma, Hepatocellular , Cell Proliferation , Gene Expression Regulation, Neoplastic , Liver Neoplasms , Neovascularization, Pathologic , RNA Stability , RNA, Messenger , Vascular Endothelial Growth Factor A , Humans , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/pathology , Carcinoma, Hepatocellular/metabolism , Liver Neoplasms/genetics , Liver Neoplasms/pathology , Liver Neoplasms/metabolism , Vascular Endothelial Growth Factor A/metabolism , Vascular Endothelial Growth Factor A/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/metabolism , Neovascularization, Pathologic/pathology , Cell Proliferation/genetics , Cell Movement/genetics , Cell Line, Tumor , Male , Female , Angiogenesis
16.
Clin Lab ; 70(4)2024 Apr 01.
Article in English | MEDLINE | ID: mdl-38623655

ABSTRACT

BACKGROUND: A typical non-neoplastic connective tissue proliferations called a pyogenic granuloma. A vascular adhesion molecule used to assess angiogenesis is the CD34 marker. The primary memberof a family of growth factors, VEGF helps in generating and maintaining the lymphatic and blood circulation systems. OBJECTIVE: The aim of the study was to know the correlation between VEGF and CD34 protein marker and pyogenic granuloma. METHODS: Thirty-one formalin fixed paraffin embedded (FFPE) blocks were taken from female pyogenic granuloma patients ranging in age from 29 to 70. The IHC was used to identify VEGF and CD34 expression in the cytoplasm of the cells. RESULTS: Seventeenout of 31 patients had VEGF positive expression. Twenty-sixout of 31 had CD34 positive expression and 5 with no expression (negative expression). Brown-stained cytoplasm showed high VEGF and CD34 expression, whereas blue stained cytoplasm showed no VEGF and CD34 expression in these cells. CONCLUSIONS: The results suggest the role of suchbiomarkers in the oral pyogenic granuloma pathogenesis, and it appears that CD34 and VEGF are valuable biomarkers in evaluating vascular and inflammatory diseases like pyogenic granuloma.


Subject(s)
Granuloma, Pyogenic , Humans , Female , Granuloma, Pyogenic/diagnosis , Granuloma, Pyogenic/etiology , Granuloma, Pyogenic/metabolism , Vascular Endothelial Growth Factor A , Vascular Cell Adhesion Molecule-1 , Neovascularization, Pathologic/complications , Neovascularization, Pathologic/metabolism , Neovascularization, Pathologic/pathology , Antigens, CD34
17.
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
18.
In Vivo ; 38(3): 1192-1198, 2024.
Article in English | MEDLINE | ID: mdl-38688651

ABSTRACT

BACKGROUND/AIM: Probing brain tumor microvasculature holds significant importance in both basic cancer research and medical practice for tracking tumor development and assessing treatment outcomes. However, few imaging methods commonly used in clinics can noninvasively monitor the brain microvascular network at high precision and without exogenous contrast agents in vivo. The present study aimed to investigate the characteristics of microvasculature during brain tumor development in an orthotopic glioma mouse model. MATERIALS AND METHODS: An orthotopic glioma mouse model was established by surgical orthotopic implantation of U87-MG-luc cells into the mouse brain. Then, optical coherence tomography angiography (OCTA) was utilized to characterize the microvasculature progression within 14 days. RESULTS: The orthotopic glioma mouse model evaluated by bioluminescence imaging and MRI was successfully generated. As the tumor grew, the microvessels within the tumor area slowly decreased, progressing from the center to the periphery for 14 days. CONCLUSION: This study highlights the potential of OCTA as a useful tool to noninvasively visualize the brain microvascular network at high precision and without any exogenous contrast agents in vivo.


Subject(s)
Brain Neoplasms , Disease Models, Animal , Glioma , Tomography, Optical Coherence , Animals , Tomography, Optical Coherence/methods , Mice , Brain Neoplasms/diagnostic imaging , Brain Neoplasms/pathology , Glioma/diagnostic imaging , Glioma/pathology , Cell Line, Tumor , Humans , Microvessels/diagnostic imaging , Microvessels/pathology , Magnetic Resonance Imaging/methods , Neovascularization, Pathologic/diagnostic imaging , Neovascularization, Pathologic/pathology , Angiography/methods
19.
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
20.
Biochem Biophys Res Commun ; 714: 149965, 2024 Jun 25.
Article in English | MEDLINE | ID: mdl-38657447

ABSTRACT

At present, the molecular mechanisms driving the progression and metastasis of oral squamous cell carcinoma (OSCC) remain largely uncharacterized. The activation of transforming growth factor-ß (TGF-ß) signaling in the tumor microenvironment has been observed in various types of cancer and has been implicated their progression by enhancing the migration and invasion of epithelial cancer cells. However, its specific roles in the oral cancer progression remain unexplored. In this study, we examined the effects of TGF-ß signaling on the murine squamous cell carcinoma, SCCVII cells in vitro and in vivo. The incubation of SCCVII cells with TGF-ß induced the activation of TGF-ß signals and epithelial-mesenchymal transition (EMT). Notably, the motility of SCCVII cells was increased upon the activation of the TGF-ß signaling. RNA sequencing revealed upregulation of genes related to EMT and angiogenesis. Consistent with these in vitro results, the inhibition of TGF-ß signals in SCCVII cell-derived primary tumors resulted in suppressed angiogenesis. Furthermore, we identified six candidate factors (ANKRD1, CCBE1, FSTL3, uPA, TSP-1 and integrin ß3), whose expression was induced by TGF-ß in SCCVII cells, and associated with poor prognosis for patients with head and neck squamous cell carcinoma. These results highlight the role of TGF-ß signals in the progression of OSCC via multiple mechanisms, including EMT and angiogenesis, and suggest novel therapeutic targets for the treatment of OSCC.


Subject(s)
Carcinoma, Squamous Cell , Disease Progression , Epithelial-Mesenchymal Transition , Neovascularization, Pathologic , Signal Transduction , Transforming Growth Factor beta , Animals , Transforming Growth Factor beta/metabolism , Carcinoma, Squamous Cell/pathology , Carcinoma, Squamous Cell/metabolism , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/blood supply , Neovascularization, Pathologic/metabolism , Neovascularization, Pathologic/pathology , Neovascularization, Pathologic/genetics , Mice , Cell Line, Tumor , Mouth Neoplasms/pathology , Mouth Neoplasms/metabolism , Mouth Neoplasms/genetics , Mouth Neoplasms/blood supply , Cell Movement/drug effects , Humans , Gene Expression Regulation, Neoplastic , Tumor Microenvironment , Angiogenesis
SELECTION OF CITATIONS
SEARCH DETAIL
...