Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 47.673
Filter
1.
Cell Death Dis ; 15(8): 563, 2024 Aug 05.
Article in English | MEDLINE | ID: mdl-39098905

ABSTRACT

Tumor neovascularization is essential for the growth, invasion, and metastasis of tumors. Recent studies have highlighted the significant role of N6-methyladenosine (m6A) modification in regulating these processes. This review explores the mechanisms by which m6A influences tumor neovascularization, focusing on its impact on angiogenesis and vasculogenic mimicry (VM). We discuss the roles of m6A writers, erasers, and readers in modulating the stability and translation of angiogenic factors like vascular endothelial growth factor (VEGF), and their involvement in key signaling pathways such as PI3K/AKT, MAPK, and Hippo. Additionally, we outline the role of m6A in vascular-immune crosstalk. Finally, we discuss the current development of m6A inhibitors and their potential applications, along with the contribution of m6A to anti-angiogenic therapy resistance. Highlighting the therapeutic potential of targeting m6A regulators, this review provides novel insights into anti-angiogenic strategies and underscores the need for further research to fully exploit m6A modulation in cancer treatment. By understanding the intricate role of m6A in tumor neovascularization, we can develop more effective therapeutic approaches to inhibit tumor growth and overcome treatment resistance. Targeting m6A offers a novel approach to interfere with the tumor's ability to manipulate its microenvironment, enhancing the efficacy of existing treatments and providing new avenues for combating cancer progression.


Subject(s)
Adenosine , Neoplasms , Neovascularization, Pathologic , Humans , Adenosine/analogs & derivatives , Adenosine/metabolism , Neovascularization, Pathologic/metabolism , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/pathology , Neoplasms/pathology , Neoplasms/metabolism , Neoplasms/genetics , Neoplasms/blood supply , Neoplasms/drug therapy , Animals , Signal Transduction
2.
J Gene Med ; 26(8): e3724, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39107869

ABSTRACT

BACKGROUND: New targeted drugs about angiogenesis could develop the treatment of glioma. We aimed to explore the role of phosducin like 3 (PDCL3) in angiogenesis of glioma. MATERIALS AND METHODS: RNA sequencing data and matched clinical data were downloaded from The Cancer Genome Atlas (TCGA) and Chinese Glioma Genome Atlas (CGGA) databases. To screen for the reliable genes with the filtering analyses, survival, multivariate Cox, receiver operating characteristic (ROC) curve filtration, and clinical correlation analyses were performed. The PDCL3 gene was validated by immunohistochemistry as a reliable gene for further analysis. Then we used the combined data of TCGA and Genotype-Tissue Expression from UCSC to detect the differential gene expression of PDCL3. Related signal pathways in glioma were explored by the gene set enrichment analysis and co-expression analysis. Lastly, we performed in vitro experiments to verify the gene functions and related mechanisms. RESULTS: The three filtering analyses and immunostaining indicated that the expression of PDCL3 in glioma tissues was higher than the normal tissues. Gene function analysis showed that PDCL3 activated the vascular endothelial growth factor (VEGF) signal pathway, and its mechanism was related to pathways in cancer, like NOD like receptor signaling pathway, the RIG-I like receptor signaling pathway and the P53 signaling pathway by MAPK/AKT in gliomas. This suggested that the proliferation, migration and invasion of glioma cells might be inhibited by the downregulation of PDCL3 in vitro, which may be related to the activation of VEGF signaling pathway. CONCLUSION: We demonstrated that PDCL3 could function as an independent adverse prognostic marker in glioma. Its pro-oncogenic mechanism may be related to the VEGF signaling pathway.


Subject(s)
Biomarkers, Tumor , Gene Expression Regulation, Neoplastic , Glioma , Signal Transduction , Vascular Endothelial Growth Factor A , Humans , Glioma/genetics , Glioma/metabolism , Glioma/pathology , Vascular Endothelial Growth Factor A/metabolism , Vascular Endothelial Growth Factor A/genetics , Prognosis , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Cell Line, Tumor , Cell Proliferation/genetics , Brain Neoplasms/genetics , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Female , Male , Cell Movement/genetics , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/metabolism , Middle Aged , Gene Expression Profiling
3.
Compr Physiol ; 14(2): 5389-5406, 2024 Mar 29.
Article in English | MEDLINE | ID: mdl-39109978

ABSTRACT

Uncontrolled angiogenesis underlies various pathological conditions such as cancer, age-related macular degeneration (AMD), and proliferative diabetic retinopathy (PDR). Hence, targeting pathological angiogenesis has become a promising strategy for the treatment of cancer and neovascular ocular diseases. However, current pharmacological treatments that target VEGF signaling have met with limited success either due to acquiring resistance against anti-VEGF therapies with serious side effects including nephrotoxicity and cardiovascular-related adverse effects in cancer patients or retinal vasculitis and intraocular inflammation after intravitreal injection in patients with AMD or PDR. Therefore, there is an urgent need to develop novel strategies which can control multiple aspects of the pathological microenvironment and regulate the process of abnormal angiogenesis. To this end, vascular normalization has been proposed as an alternative for antiangiogenesis approach; however, these strategies still focus on targeting VEGF or FGF or PDGF which has shown adverse effects. In addition to these growth factors, calcium has been recently implicated as an important modulator of tumor angiogenesis. This article provides an overview on the role of major calcium channels in endothelium, TRP channels, with a special focus on TRPV4 and its downstream signaling pathways in the regulation of pathological angiogenesis and vascular normalization. We also highlight recent findings on the modulation of TRPV4 activity and endothelial phenotypic transformation by tumor microenvironment through Rho/YAP/VEGFR2 mechanotranscriptional pathways. Finally, we provide perspective on endothelial TRPV4 as a novel VEGF alternative therapeutic target for vascular normalization and improved therapy. © 2024 American Physiological Society. Compr Physiol 14:5389-5406, 2024.


Subject(s)
Neovascularization, Pathologic , Humans , Animals , Transient Receptor Potential Channels/metabolism , Transient Receptor Potential Channels/physiology , Signal Transduction
4.
Med Oncol ; 41(9): 217, 2024 Aug 05.
Article in English | MEDLINE | ID: mdl-39102060

ABSTRACT

Multiple myeloma (MM) is a neoplastic condition resulting from the uncontrolled expansion of B-cell-derived plasma cells. The importance of angiogenesis in MM development has also been demonstrated. Extracellular vesicles (EVs) have vital functions in interactions between neighboring cells, such as angiogenesis. The objective of this in vitro study was to examine the transfection and angiogenesis effects of MM-EVs on endothelial cells (ECs) upon treatment with Tetrahydroisoquinoline (THIQ) as a bioactive organic compound derivative from isoquinoline. Following treatment of multiple myeloma cells (U266) with THIQ, MM-EVs were harvested and transmigrated to human umbilical vein endothelial cells (HUVEC) in a co-culture model. EVs transmigration was traced by flow cytometry. Correspondingly, the expression of angiogenic genes and/or proteins in U266 cells and HUVECs was measured by RT-PCR and ELISA methods. Likewise, the proliferation and migration of HUVECs treated with THIQ-treated MM-EVs were visualized and estimated by performing both tube formation and scratch wound healing methods. Surprisingly, the anti-angiogenic effect of THIQ-treated MM-EVs was evident by the decreased expression of CD34, VEGFR2, and IL-6 at the mRNA and/or protein levels after internalization of MM-EVs in HUVEC. Finally, tube formation and scratch wound healing experiments showed inhibition of HUVEC cell proliferation and migration by THIQ-treated MM-EVs compared to control MM-EVs. MM-EVs derived from THIQ-treated myeloma cells (U266) inhibited angiogenesis in HUVECs. This phenomenon is coordinated by the internalized THIQ-treated MM-EVs in HUVECs, and ultimately the reduction of angiogenic factors and inhibition of tube formation and scratch wound healing.


Subject(s)
Cell Movement , Extracellular Vesicles , Human Umbilical Vein Endothelial Cells , Multiple Myeloma , Neovascularization, Pathologic , Tetrahydroisoquinolines , Humans , Human Umbilical Vein Endothelial Cells/drug effects , Human Umbilical Vein Endothelial Cells/metabolism , Multiple Myeloma/pathology , Multiple Myeloma/drug therapy , Multiple Myeloma/metabolism , Extracellular Vesicles/metabolism , Extracellular Vesicles/drug effects , Neovascularization, Pathologic/drug therapy , Neovascularization, Pathologic/pathology , Neovascularization, Pathologic/metabolism , Tetrahydroisoquinolines/pharmacology , Cell Movement/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Coculture Techniques , Angiogenesis Inhibitors/pharmacology , Angiogenesis
5.
J Pathol Clin Res ; 10(5): e12393, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39119942

ABSTRACT

Angiogenesis is recognized as a hallmark of cancer, and vascular endothelial growth factor (VEGF) is a key regulator of the angiogenic process and is related to cancer progression. Anti-VEGF therapy has been tried but with limited success and without useful stratification for angiogenesis markers. Further, the landscape of VEGF single nucleotide polymorphisms (SNPs) in breast cancer and their clinical relevance is not well studied, and their relation to tissue-based angiogenesis markers has not been explored. Here, we studied a selection of VEGFA SNPs in nontumor lymph nodes from a population-based breast cancer cohort (n = 544), and their relation to clinicopathologic variables, vascular tissue metrics, and breast cancer-specific survival. Two of the SNP candidates (rs833068GA genotype and rs25648CC genotype) showed associations with angiogenesis tissue markers, and the VEGFA rs833068GA genotype was associated with breast cancer-specific survival among ER-negative cases. We also found trends of association between the rs699947CA genotype and large tumor diameter and ER-negative tumors, and between the rs3025039CC genotype and large tumor diameter. Our findings indicate some associations between certain VEGF SNPs, in particular the rs833068GA genotype, and both vascular metrics and patient survival. These findings and their potential implications need to be validated by independent studies.


Subject(s)
Breast Neoplasms , Disease Progression , Polymorphism, Single Nucleotide , Vascular Endothelial Growth Factor A , Humans , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Female , Vascular Endothelial Growth Factor A/genetics , Middle Aged , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/pathology , Biomarkers, Tumor/genetics , Aged , Adult , Genotype , Genetic Predisposition to Disease
6.
Cells ; 13(15)2024 Jul 24.
Article in English | MEDLINE | ID: mdl-39120274

ABSTRACT

MicroRNAs (miRs) regulate physiological and pathological processes, including ischemia-induced angiogenesis and neovascularization. They can be transferred between cells by extracellular vesicles (EVs). However, the specific miRs that are packaged in EVs released from skeletal muscles, and how this process is modulated by ischemia, remain to be determined. We used a mouse model of hindlimb ischemia and next generation sequencing (NGS) to perform a complete profiling of miR expression and determine the effect of ischemia in skeletal muscles, and in EVs of different sizes (microvesicles (MVs) and exosomes) released from these muscles. Ischemia significantly modulated miR expression in whole muscles and EVs, increasing the levels of several miRs that can have pro-angiogenic effects (angiomiRs). We found that specific angiomiRs are selectively enriched in MVs and/or exosomes in response to ischemia. In silico approaches indicate that these miRs modulate pathways that play key roles in angiogenesis and neovascularization, including HIF1/VEGF signaling, regulation of actin cytoskeleton and focal adhesion, NOTCH, PI3K/AKT, RAS/MAPK, JAK/STAT, TGFb/SMAD signaling and the NO/cGMP/PKG pathway. Thus, we show for the first time that angiomiRs are selectively enriched in MVs and exosomes released from ischemic muscles. These angiomiRs could be targeted in order to improve the angiogenic function of EVs for potential novel therapeutic applications in patients with severe ischemic vascular diseases.


Subject(s)
Extracellular Vesicles , Ischemia , MicroRNAs , Muscle, Skeletal , Neovascularization, Physiologic , Animals , Muscle, Skeletal/metabolism , Muscle, Skeletal/blood supply , Muscle, Skeletal/pathology , Extracellular Vesicles/metabolism , MicroRNAs/metabolism , MicroRNAs/genetics , Ischemia/metabolism , Ischemia/pathology , Mice , Hindlimb/blood supply , Hindlimb/pathology , Mice, Inbred C57BL , Signal Transduction , Male , Exosomes/metabolism , Neovascularization, Pathologic/metabolism , Angiogenesis
7.
Cells ; 13(15)2024 Jul 31.
Article in English | MEDLINE | ID: mdl-39120324

ABSTRACT

High levels of vascular endothelial growth factor (VEGF), fibroblast growth factor (FGF)-2 and angiopoietin (ANG)-2 are found in tissues from oral squamous cell carcinoma (OSCC) and oral potentially malignant disorders (OPMDs). As might be expected, VEGF, FGF-2, and ANG-2 overexpression parallels the development of new blood and lymphatic vessels that nourish the growing OPMDs or OSCCs and provide the latter with metastatic routes. Notably, VEGF, FGF-2, and ANG-2 are also linked to the epithelial-to-mesenchymal transition (EMT), a trans-differentiation process that respectively promotes or exasperates the invasiveness of normal and neoplastic oral epithelial cells. Here, we have summarized published work regarding the impact that the interplay among VEGF, FGF-2, ANG-2, vessel generation, and EMT has on oral carcinogenesis. Results from the reviewed studies indicate that VEGF, FGF-2, and ANG-2 spark either protein kinase B (AKT) or mitogen-activated protein kinases (MAPK), two signaling pathways that can promote both EMT and new vessels' formation in OPMDs and OSCCs. Since EMT and vessel generation are key to the onset and progression of OSCC, as well as to its radio- and chemo-resistance, these data encourage including AKT or MAPK inhibitors and/or antiangiogenic drugs in the treatment of this malignancy.


Subject(s)
Carcinoma, Squamous Cell , Epithelial-Mesenchymal Transition , Mouth Neoplasms , Humans , Mouth Neoplasms/pathology , Mouth Neoplasms/metabolism , Carcinoma, Squamous Cell/pathology , Carcinoma, Squamous Cell/metabolism , Disease Progression , Vascular Endothelial Growth Factor A/metabolism , Neovascularization, Pathologic/metabolism , Neovascularization, Pathologic/pathology , Animals , Angiogenesis Inducing Agents/metabolism , Fibroblast Growth Factor 2/metabolism , Signal Transduction
8.
Clin Exp Med ; 24(1): 178, 2024 Aug 06.
Article in English | MEDLINE | ID: mdl-39105831

ABSTRACT

This review explores the pivotal role of angiogenesis in breast cancer progression and treatment. It covers biomarkers, imaging techniques, therapeutic approaches, resistance mechanisms, and clinical implications. Key topics include Vascular Endothelial Growth Factors, angiopoietins, microRNA signatures, and circulating endothelial cells as biomarkers, along with Magnetic Resonance Imaging, Computed Tomography Angiography, Ultrasound, and Positron Emission Tomography for imaging. Therapeutic strategies targeting VEGF, tyrosine kinase inhibitors, and the intersection of angiogenesis with immunotherapy are discussed. Challenges such as resistance mechanisms and personalized medicine approaches are addressed. Clinical implications, prognostic value, and the future direction of angiogenesis-targeted therapies are highlighted. The article concludes with reflections on the transformative potential of understanding angiogenesis.


Subject(s)
Breast Neoplasms , Neovascularization, Pathologic , Female , Humans , Angiogenesis/drug effects , Angiogenesis/physiopathology , Angiogenesis Inhibitors/therapeutic use , Breast Neoplasms/pathology , Neovascularization, Pathologic/drug therapy , Neovascularization, Pathologic/physiopathology
9.
Immun Inflamm Dis ; 12(8): e1370, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39110084

ABSTRACT

BACKGROUND: Endometriosis is associated with a wide variety of signs and symptoms and can lead to infertility, embryo death, and even miscarriage. Although the exact pathogenesis and etiology of endometriosis is still unclear, it has been shown that it has a chronic inflammatory nature and angiogenesis is also involved in it. OBJECTIVE: This review aims to explore the role of inflammation and angiogenesis in endometriosis and suggest a potential treatment targeting these pathways. FINDINGS: Among the pro-inflammatory cytokines, studies have shown solid roles for interleukin 1ß (IL-ß), IL-6, and tumor necrosis factor α (TNF-α) in the pathogenesis of this condition. Other than inflammation, angiogenesis, the formation of new blood vessels from pre-existing capillaries, is also involved in the pathogenesis of endometriosis. Among angiogenic factors, vascular endothelial growth factor (VEGF), hypoxia-inducible factor 1α (HIF-1α), transforming growth factor ß1 (TGF-ß1), and matrix metalloproteinases (MMPs) are more essential in the pathogenesis of endometriosis. Interestingly, it has been shown that inflammation and angiogenesis share some similar pathways with each other that could be potentially targeted for treatment of diseases caused by these two processes. Cannabidiol (CBD) is a non-psychoactive member of cannabinoids which has well-known and notable anti-inflammatory and antiangiogenic properties. This agent has been shown to decrease IL-1ß, IL-6, TNF-α, VEGF, TGFß, and MMPs in different animal models of diseases. CONCLUSION: It seems that CBD could be a possible treatment for endometriosis due to its anti-inflammatory and antiangiogenic activity, however, further studies are needed.


Subject(s)
Cannabidiol , Endometriosis , Inflammation , Neovascularization, Pathologic , Endometriosis/drug therapy , Endometriosis/pathology , Female , Humans , Cannabidiol/therapeutic use , Cannabidiol/pharmacology , Neovascularization, Pathologic/drug therapy , Inflammation/drug therapy , Animals , Cytokines/metabolism , Anti-Inflammatory Agents/therapeutic use , Anti-Inflammatory Agents/pharmacology , Angiogenesis Inhibitors/therapeutic use , Angiogenesis Inhibitors/pharmacology , Angiogenesis
10.
Technol Cancer Res Treat ; 23: 15330338241273239, 2024.
Article in English | MEDLINE | ID: mdl-39110070

ABSTRACT

Tumor growth and metastasis rely on angiogenesis. In recent years, long non-coding RNAs have been shown to play an important role in regulating tumor angiogenesis. Here, we review the multidimensional modes and relevant molecular mechanisms of long non-coding RNAs in regulating tumor angiogenesis. In addition, we summarize new strategies for tumor anti-angiogenesis therapies by targeting long non-coding RNAs. The aim of this study is to provide new diagnostic targets and treatment strategies for anti-angiogenic tumor therapy.


Subject(s)
Gene Expression Regulation, Neoplastic , Neoplasms , Neovascularization, Pathologic , RNA, Long Noncoding , RNA, Long Noncoding/genetics , Humans , Neovascularization, Pathologic/genetics , Neoplasms/genetics , Neoplasms/pathology , Neoplasms/therapy , Neoplasms/blood supply , Animals , Angiogenesis Inhibitors/therapeutic use , Molecular Targeted Therapy , Angiogenesis
12.
Eur J Histochem ; 68(3)2024 Aug 06.
Article in English | MEDLINE | ID: mdl-39105608

ABSTRACT

Endometriosis is an estrogen-dependent inflammatory gynecological disease whose pathogenesis is unclear. C-C motif chemokine ligand 18 (CCL18), a chemokine, is involved in several inflammatory diseases. In this study, we aimed to investigate the role of CCL18 in endometriosis and its underlying mechanisms. Human endometrium and peritoneal fluid were obtained from women with and without endometriosis for molecular studies. The expression level of CCL18 in each tissue sample was examined by RNA sequencing analysis, quantitative PCR analysis and immunohistochemistry staining. The effects of CCL18 on cell migration, tube formation and neurite growth were investigated in vitro using primary endometrial cells, human umbilical vein endothelial cells (HUVECs) and dorsal root ganglion (DRG) neurons, respectively. Moreover, the development of endometriosis in mice was studied in vivo by blocking CCL18. CCL18 was shown to be overexpressed in endometrial foci and peritoneal fluid in women with endometriosis and was positively correlated with endometriosis pain. In vitro, CCL18 promoted the migration of ectopic endometrial cells, tube formation of HUVECs, and nerve outgrowth of DRG neurons. More importantly, inhibition of CCL18 significantly suppressed lesion development, angiogenesis, and nerve infiltration in a mouse model of endometriosis. In conclusion, CCL18 may play a role in the progression of endometriosis by increasing endometrial cell migration and promoting neuroangiogenesis.


Subject(s)
Cell Movement , Chemokines, CC , Endometriosis , Endometrium , Human Umbilical Vein Endothelial Cells , Neovascularization, Pathologic , Endometriosis/metabolism , Endometriosis/pathology , Female , Humans , Animals , Endometrium/metabolism , Endometrium/pathology , Human Umbilical Vein Endothelial Cells/metabolism , Mice , Neovascularization, Pathologic/metabolism , Neovascularization, Pathologic/pathology , Chemokines, CC/metabolism , Adult , Ganglia, Spinal/metabolism , Ganglia, Spinal/pathology , Ascitic Fluid/metabolism , Ascitic Fluid/pathology , Mice, Inbred C57BL
13.
Braz J Med Biol Res ; 57: e13889, 2024.
Article in English | MEDLINE | ID: mdl-39194034

ABSTRACT

With the escalating incidence and mortality rates of cancer, there is an ever-growing emphasis on the research of anticancer drugs. Cordycepin, the primary nucleoside antibiotic isolated from Cordyceps militaris, has emerged as a remarkable agent for cancer prevention and treatment. Functioning as a natural targeted antitumor drug, cordycepin assumes an increasingly pivotal role in cancer therapy. This review elucidates the mechanisms of cordycepin in inhibiting tumor cell proliferation, inducing apoptosis, as well as its capabilities in suppressing angiogenesis and metastasis. Moreover, the immunomodulatory effects of cordycepin in cancer treatment are explored. Additionally, the current status, challenges, and future prospects of cordycepin application in clinical trials are briefly discussed. The objective is to provide a valuable reference for the utilization of cordycepin in cancer treatment.


Subject(s)
Apoptosis , Cell Proliferation , Deoxyadenosines , Neoplasms , Deoxyadenosines/pharmacology , Deoxyadenosines/therapeutic use , Humans , Neoplasms/drug therapy , Cell Proliferation/drug effects , Apoptosis/drug effects , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Neovascularization, Pathologic/drug therapy
14.
Cells ; 13(16)2024 Aug 12.
Article in English | MEDLINE | ID: mdl-39195226

ABSTRACT

Ectosomes are carriers of proangiogenic factors during cancer progression. This study investigated whether the proangiogenic effect exerted by melanoma-derived ectosomes on recipient endothelial cells is mediated by ectosomal αvß3 and αvß5 integrins. Ectosomes were isolated from the conditioned culture media of four melanoma cell lines and melanocytes. Changes in gene and protein expression of αvß3 and αvß5 integrins, as well as VEGF and TNF-α were assessed in ectosome-treated endothelial cells. To confirm the functional involvement of ectosomal integrins in functional tests (Alamar Blue, wound healing and tube formation assays), ectosomes were also pretreated with anti-integrin antibodies and integrin-blocking peptides echistatin and cilengitide. Melanoma-derived ectosomes induced changes in the expression of αvß3 and αvß5 integrins in recipient endothelial cells, leading to increased viability, migratory properties, and tube formation potential. The extent of proangiogenic stimulation varied depending on the types of cells releasing ectosomes and the recipient cells. The use of anti-integrin antibodies and integrin-blocking peptides revealed a more significant role for the αvß5 integrin/VEGF than the αvß3 integrin/TNF-α pathway in the interactions between ectosomes and endothelial cells. The study demonstrated the functional role of ectosomal αvß3 and αvß5 integrins. It also provided a baseline understanding of ectosome-mediated αvß3 integrin/TNF-α and αvß5 integrin/VEGF signaling in angiogenesis.


Subject(s)
Integrin alphaVbeta3 , Melanoma , Receptors, Vitronectin , Humans , Melanoma/pathology , Melanoma/metabolism , Integrin alphaVbeta3/metabolism , Receptors, Vitronectin/metabolism , Cell Line, Tumor , Cell Movement/drug effects , Vascular Endothelial Growth Factor A/metabolism , Tumor Necrosis Factor-alpha/metabolism , Neovascularization, Pathologic/metabolism , Endothelial Cells/metabolism , Endothelial Cells/drug effects , Human Umbilical Vein Endothelial Cells/metabolism
15.
Sci Rep ; 14(1): 18363, 2024 08 07.
Article in English | MEDLINE | ID: mdl-39112724

ABSTRACT

The combination of anti-angiogenic treatment and immunotherapy presents a promising strategy against colon cancer. Interleukin-17F (IL-17F) emerges as a critical immune cell cytokine expressed in colonic epithelial cells, demonstrating potential in inhibiting angiogenesis. In order to clarify the roles of IL-17F in the colon cancer microenvironment and elucidate its mechanism, we established a mouse colon carcinoma cell line CT26 overexpressing IL-17F and transplanted it subcutaneously into syngeneic BALB/c mice. We also analyzed induced colon tumor in IL-17F knockout and wild type mice. Our results demonstrated that IL-17F could suppress colon tumor growth in vivo with inhibited angiogenesis and enhanced recruitment of cysteine-cysteine motif chemokine receptor 6 (CCR6) positive immune cells. Additionally, IL-17F suppressed the tube formation, cell growth and migration of endothelial cells EOMA in vitro. Comprehensive bioinformatics analysis of transcriptome profiles between EOMA cells and those treated with three different concentrations of IL-17F identified 109 differentially expressed genes. Notably, a potential new target, Caspase 4, showed increased expressions after IL-17F treatment in endothelial cells. Further molecular validation revealed a novel downstream signaling for IL-17F: IL-17F enhanced Caspase 4/GSDMD signaling of endothelial cells, CT26 cells and CT26 transplanted tumors, while IL-17F knockout colon tumors exhibited decreased Caspase 4/GSDMD signaling. The heightened expression of the GSDMD N-terminus, coupled with increased cellular propidium iodide (PI) uptake and lactate dehydrogenase (LDH) release, revealed that IL-17F promoted pyroptosis of endothelial cells. Altogether, IL-17F could modulate the colon tumor microenvironment with inhibited angiogenesis, underscoring its potential as a therapeutic target for colon cancer.


Subject(s)
Colonic Neoplasms , Endothelial Cells , Interleukin-17 , Mice, Inbred BALB C , Pyroptosis , Animals , Colonic Neoplasms/pathology , Colonic Neoplasms/metabolism , Colonic Neoplasms/genetics , Interleukin-17/metabolism , Mice , Endothelial Cells/metabolism , Cell Line, Tumor , Caspases, Initiator/metabolism , Caspases, Initiator/genetics , Neovascularization, Pathologic/metabolism , Neovascularization, Pathologic/genetics , Mice, Knockout , Tumor Microenvironment , Humans , Cell Proliferation
16.
Front Immunol ; 15: 1414450, 2024.
Article in English | MEDLINE | ID: mdl-39165361

ABSTRACT

Background: In the ongoing battle against breast cancer, a leading cause of cancer-related mortality among women globally, the urgent need for innovative prognostic markers and therapeutic targets is undeniable. This study pioneers an advanced methodology by integrating machine learning techniques to unveil a vascular mimicry signature, offering predictive insights into breast cancer outcomes. Vascular mimicry refers to the phenomenon where cancer cells mimic blood vessel formation absent of endothelial cells, a trait associated with heightened tumor aggression and diminished response to conventional treatments. Methods: The study's comprehensive analysis spanned data from over 6,000 breast cancer patients across 12 distinct datasets, incorporating both proprietary clinical data and single-cell data from 7 patients, accounting for a total of 43,095 cells. By employing an integrative strategy that utilized 10 machine learning algorithms across 108 unique combinations, the research scrutinized 100 existing breast cancer signatures. Empirical validation was sought through immunohistochemistry assays, alongside explorations into potential immunotherapeutic and chemotherapeutic avenues. Results: The investigation successfully identified six genes related to vascular mimicry from multi-center cohorts, laying the groundwork for a novel predictive model. This model outstripped the prognostic accuracy of traditional clinical and molecular indicators in forecasting recurrence and mortality risks. High-risk individuals identified by our model faced worse outcomes. Further validation through IHC assays in 30 patients underscored the model's extensive applicability. Notably, the model unveiled varying therapeutic responses; low-risk patients might achieve greater benefits from immunotherapy, whereas high-risk patients demonstrated a particular sensitivity to certain chemotherapies, such as ispinesib. Conclusions: This model marks a significant step forward in the precise evaluation of breast cancer prognosis and therapeutic responses across different patient groups. It heralds the possibility of refining patient outcomes through tailored treatment strategies, accentuating the potential of machine learning in revolutionizing cancer prognosis and management.


Subject(s)
Breast Neoplasms , Machine Learning , Humans , Female , Breast Neoplasms/pathology , Prognosis , Biomarkers, Tumor , Biological Mimicry , Neovascularization, Pathologic
17.
Sci Transl Med ; 16(761): eadn0904, 2024 Aug 21.
Article in English | MEDLINE | ID: mdl-39167664

ABSTRACT

Deficit of oxygen and nutrients in the tumor microenvironment (TME) triggers abnormal angiogenesis that produces dysfunctional and leaky blood vessels, which fail to adequately perfuse tumor tissues. Resulting hypoxia, exacerbation of metabolic disturbances, and generation of an immunosuppressive TME undermine the efficacy of anticancer therapies. Use of carefully scheduled angiogenesis inhibitors has been suggested to overcome these problems and normalize the TME. Here, we propose an alternative agonist-based normalization approach using a derivative of the C-type natriuretic peptide (dCNP). Multiple gene expression signatures in tumor tissues were affected in mice treated with dCNP. In several mouse orthotopic and subcutaneous solid tumor models including colon and pancreatic adenocarcinomas, this well-tolerated agent stimulated formation of highly functional tumor blood vessels to reduce hypoxia. Administration of dCNP also inhibited stromagenesis and remodeling of the extracellular matrix and decreased tumor interstitial fluid pressure. In addition, treatment with dCNP reinvigorated the antitumor immune responses. Administration of dCNP decelerated growth of primary mouse tumors and suppressed their metastases. Moreover, inclusion of dCNP into the chemo-, radio-, or immune-therapeutic regimens increased their efficacy against solid tumors in immunocompetent mice. These results demonstrate the proof of principle for using vasculature normalizing agonists to improve therapies against solid tumors and characterize dCNP as the first in class among such agents.


Subject(s)
Natriuretic Peptide, C-Type , Neovascularization, Pathologic , Tumor Microenvironment , Animals , Neovascularization, Pathologic/drug therapy , Natriuretic Peptide, C-Type/pharmacology , Natriuretic Peptide, C-Type/therapeutic use , Mice , Humans , Tumor Microenvironment/drug effects , Cell Line, Tumor , Neoplasms/drug therapy , Neoplasms/pathology , Neoplasms/blood supply , Immunity/drug effects
18.
BMC Cancer ; 24(1): 1040, 2024 Aug 22.
Article in English | MEDLINE | ID: mdl-39174921

ABSTRACT

BACKGROUND: Glioblastoma (GBM) is a malignant astrocytic tumor and its progression involves the regulation of vascular endothelial growth factor-A (VEGFA). However, the mechanism of VEGFA in regulating GBM progression remains unclear. METHODS: VEGFA mRNA expression was analyzed by quantitative real-time polymerase chain reaction. Protein expression of VEGFA, cluster of differentiation 9 (CD9), CD81, and transforming growth factor-ß1 (TGF-ß1) was detected by western blotting assay. Flow cytometry assay was conducted to assess cell proliferation, cell apoptosis and myeloid-derived suppressor cell (MDSC) differentiation. TUNEL cell apoptosis detection kit was utilized to analyze cell apoptosis of tumors. Angiogenic capacity was investigated by tube formation assay. Transwell assay was used to assess cell migration and invasion. The effect of VEGFA on tumor formation was determined by a xenograft mouse model assay. Immunohistochemistry assay was used to analyze positive expression rate of VEGFA in tumor tissues. TGF-ß1 level was detected by enzyme-linked immunosorbent assay. RESULTS: VEGFA expression was upregulated in GBM tissues, GBM cells, and exosomes from GBM patients and GBM cells. VEGFA silencing led to decreased cell proliferation, tube formation, migration and invasion and increased cell apoptosis. Moreover, VEGFA knockdown also delayed tumor formation. VEGFA promoted MDSC differentiation and TGF-ß1 secretion by MDSCs by being packaged into exosomes. In addition, TGF-ß1 knockdown displayed similar effects with VEGFA silencing on GBM cell phenotypes, and MDSCs attenuated VEGFA knockdown-induced effects by secreting TGF-ß1 in A172 and U251 cells. CONCLUSION: VEGFA contributed to tumor property of GBM cells by promoting MDSC differentiation and TGF-ß1 secretion by MDSCs, providing potential targets for GBM treatment.


Subject(s)
Apoptosis , Cell Differentiation , Cell Proliferation , Glioblastoma , Myeloid-Derived Suppressor Cells , Transforming Growth Factor beta1 , Vascular Endothelial Growth Factor A , Glioblastoma/pathology , Glioblastoma/metabolism , Glioblastoma/genetics , Humans , Animals , Vascular Endothelial Growth Factor A/metabolism , Vascular Endothelial Growth Factor A/genetics , Mice , Myeloid-Derived Suppressor Cells/metabolism , Myeloid-Derived Suppressor Cells/pathology , Transforming Growth Factor beta1/metabolism , Transforming Growth Factor beta1/genetics , Cell Line, Tumor , Cell Movement/genetics , Brain Neoplasms/pathology , Brain Neoplasms/metabolism , Brain Neoplasms/genetics , Gene Expression Regulation, Neoplastic , Neovascularization, Pathologic/metabolism , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/pathology , Xenograft Model Antitumor Assays , Female
19.
Crit Rev Eukaryot Gene Expr ; 34(8): 25-34, 2024.
Article in English | MEDLINE | ID: mdl-39180205

ABSTRACT

Lysine demethylase 6A (KDM6A) is abnormally expressed in various cancer. This study aimed to investigate the potential of KDM6A in pancreatic cancer (PC). mRNA expression was calculated by reverse transcription quantitative real-time polymerase chain reaction (RT-qPCR). Protein expression was detected by Western blot. Cell viability was measured by Cell Counting Kit (CCK-8) assay. Cell angiogenesis was determined by tube formation assay. Cell migration and invasion were determined by Transwell assay. We found that KDM6A was upregulated in PC patients and cells. Interestingly, KDM6A deficiency inhibited the proliferation and angiogenesis of PC cells. Moreover, KDM6A knockdown suppressed the migration and invasion of PC cells. Additionally, KDM6A upregulated the expression of lysosomal associated membrane protein 3 (LAMP3) via driving demethylation of H3K27me3. Overexpression of LAMP3 reversed the effects of KDM6A knockdown and contributed to the angiogenesis and aggressiveness of PC cells. In summary, KDM6A-mediated demethylation of tri-methylation at lysine 27 of histone H3 (H3K27me3) promotes the transcription of LAMP3, resulting the angiogenesis and aggressiveness of PC. Therefore, targeting KDM6A may be an anti-angiogenetic strategy for PC.


Subject(s)
Cell Movement , Cell Proliferation , Gene Expression Regulation, Neoplastic , Histone Demethylases , Lysosomal Membrane Proteins , Neoplasm Invasiveness , Neovascularization, Pathologic , Pancreatic Neoplasms , Humans , Pancreatic Neoplasms/pathology , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/metabolism , Cell Movement/genetics , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/metabolism , Histone Demethylases/metabolism , Histone Demethylases/genetics , Cell Proliferation/genetics , Cell Line, Tumor , Lysosomal Membrane Proteins/metabolism , Lysosomal Membrane Proteins/genetics , Angiogenesis , Neoplasm Proteins , Lysosomal-Associated Membrane Protein 3
20.
Cell Signal ; 122: 111346, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39147296

ABSTRACT

BACKGROUND: Eplerenone is a selective aldosterone receptor blocker that is effective in preventing the progression of chroinic kidney disease (CKD). However, its mechanism and role in CKD pregnancy still remain uncertain. The aim of this study was to evaluate whether eplerenone could attenuated the fibrosis of unilateral ureteral obstruction (UUO) pregnant rats' contralateral kidney, improved pregnancy outcome and explore its therapeutic mechanisms. METHODS: A pregnancy rat model of UUO established, female Wistar rats were randomly assigned into sham-operated group (Sham group),sham-operated combined pregnancy group (SP group), unilateral ureteral obstruction combined pregnancy group (UUO + Pregnancy group), unilateral ureteral obstruction combined pregnancy, administered eplerenone (UUO + Pregnancy+Eplerenone group). On the 18th day of pregnancy, the rats were placed in a metabolic cage, 24 h urine was collected and stored at -80 °C. Next day, all animals were euthanized, and serum was collected by centrifugation and stored at -20 °C. Then the right kidney was extracted, a part of the kidney was placed in 4% paraformaldehyde for morphology, immunohistochemical staining, and immunofluorescence staining, and the other part was placed in a - 80 °C refrigerator for RNA and protein extraction. In vitro, HUVECs was treated with aldosterone, progesterone and estradiol, VEGFA and its receptor blocker bevacizumab. The ability of proliferation, migration and tubularization of HUVECs was detected by CCK-8, scratch wound assay and endothelial tube formation assay. And the co-expression of CD34 and α-SMA of HUVECs was detected by Flow cytometry. RESULTS: Immunofluorescence results showed that the co-expression of CD34 and α-SMA increased in the UUO + Pregnancy group was significantly increased. The expression of SGK-1, TGFß-1, Smad2, Smad3, VEGF-A, VEGFR2, CD34, α-SMA and Collagen I was significantly higher in the kidneys of the UUO + Pregnancy group compared to the Sham group and SP group. Eplerenone inhibited the expression of those results. In vitro, the ability of proliferation, migration and tubularization was increased after treated with aldosterone, aldosterone with progesterone and estradiol or VEGFA. Similarly, the expression of α-SMA on the surface of HUVECs treated with aldosterone, aldosterone with progesterone and estradiol were increased, while eplerenone supressed its expression. CONCLUSION: Eplerenone inhibits renal angiogenesis by blocking the SGK-1/TGFß signal transduction pathway, thereby inhibiting the phenotypic transformation of endothelial cells, slowing down renal fibrosis, and reducing kidney damage caused by pregnancy.


Subject(s)
Eplerenone , Immediate-Early Proteins , Kidney , Protein Serine-Threonine Kinases , Rats, Wistar , Renal Insufficiency, Chronic , Transforming Growth Factor beta , Animals , Female , Pregnancy , Eplerenone/pharmacology , Eplerenone/therapeutic use , Rats , Immediate-Early Proteins/metabolism , Protein Serine-Threonine Kinases/metabolism , Renal Insufficiency, Chronic/metabolism , Renal Insufficiency, Chronic/drug therapy , Renal Insufficiency, Chronic/pathology , Kidney/metabolism , Kidney/pathology , Kidney/drug effects , Transforming Growth Factor beta/metabolism , Signal Transduction/drug effects , Humans , Human Umbilical Vein Endothelial Cells/metabolism , Cell Proliferation/drug effects , Spironolactone/pharmacology , Spironolactone/analogs & derivatives , Spironolactone/therapeutic use , Ureteral Obstruction/metabolism , Ureteral Obstruction/drug therapy , Ureteral Obstruction/pathology , Ureteral Obstruction/complications , Mineralocorticoid Receptor Antagonists/pharmacology , Mineralocorticoid Receptor Antagonists/therapeutic use , Vascular Endothelial Growth Factor A/metabolism , Cell Movement/drug effects , Neovascularization, Pathologic/metabolism , Neovascularization, Pathologic/drug therapy , Angiogenesis
SELECTION OF CITATIONS
SEARCH DETAIL