Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 51
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Int J Mol Med ; 53(4)2024 04.
Article in English | MEDLINE | ID: mdl-38391009

ABSTRACT

Heart disease remains a global health challenge, contributing notably to morbidity and mortality. The lymphatic vasculature, an integral component of the cardiovascular system, plays a crucial role in regulating essential physiological processes, including fluid balance, transportation of extravasated proteins and immune cell trafficking, all of which are important for heart function. Through thorough scientometric analysis and extensive research, the present review identified lymphangiogenesis as a hotspot in cardiovascular disease research, and the mechanisms underlying impaired cardiac lymphangiogenesis and inadequate lymph drainage in various cardiovascular diseases are discussed. Furthermore, the way used to improve lymphangiogenesis to effectively regulate a variety of heart diseases and associated signaling pathways was investigated. Notably, the current review also highlights the impact of Traditional Chinese Medicine (TCM) on lymphangiogenesis, aiming to establish a clinical basis for the potential of TCM to improve cardiovascular diseases by promoting lymphangiogenesis.


Subject(s)
Cardiovascular Diseases , Heart Diseases , Lymphatic Vessels , Humans , Lymphangiogenesis/physiology , Cardiovascular Diseases/metabolism , Lymphatic Vessels/metabolism , Heart Diseases/metabolism , Heart
2.
PLoS One ; 18(12): e0295836, 2023.
Article in English | MEDLINE | ID: mdl-38100487

ABSTRACT

The establishment of new connections after NVLNT (non-vascularized lymph node transplantation) is still poorly understood. The purpose of this study was to investigate lymphatic connections after NVLNT using lymphangiography. In a mice model, 40 mice were allocated to undergo NVLNT or sham surgery. On day 21 after NVLNT, the lymphatic vessels were observed on near-infrared fluorescence imaging with indocyanine green. In a minipig model, 12 minipigs underwent NVLNT. On day 14 after NVLNT, the transplanted lymph node and donor site were checked by ultrasound, and minipigs with viable transplanted LNs were allocated to lipiodol lymphangiography or MR lymphangiography groups. Transplanted LN engraftment was examined with immunohistochemical staining. After NVLNT in mice, the signal intensities in the popliteal region at 3 minutes and 5 minutes were higher in the transplanted side than the control side (21.3 ± 8.1 vs. 11.0 ± 4.6 at 3 minutes, 26.7 ± 6.8 vs. 19.7 ± 5.9 at 5 minutes), while in the sham group, there were no significant differences between sides. In minipigs, lipiodol lymphangiography (n = 5) showed Lipiodol accumulation in transplanted LNs with innumerable newly formed lymphatic vessels and lymphovenous shunts. MR lymphangiography (n = 5) showed higher enhancement on the transplanted side compared to the control side. Histology showed successful engraftment of transplanted LNs in 16 out of 20 (80%) mice and 9 out of 12 (75%) minipigs. Omnidirectional lymphangiogenesis forming a dense lymphatic network and spontaneous formation of lymphovenous shunts were shown after NVLNT.


Subject(s)
Lymphatic Vessels , Lymphography , Swine , Animals , Mice , Lymphography/methods , Lymphangiogenesis , Ethiodized Oil , Swine, Miniature , Lymph Nodes/diagnostic imaging , Lymph Nodes/pathology , Lymphatic Vessels/diagnostic imaging , Lymphatic Vessels/pathology
3.
Phytomedicine ; 118: 154926, 2023 Sep.
Article in English | MEDLINE | ID: mdl-37392675

ABSTRACT

BACKGROUND: Lymph node metastasis is a key mechanism in gastric cancer (GC) metastasis and lymphangiogenesis is a vital step in the process of lymph node metastasis. Currently, there are no drugs which can treat lymph node metastasis in GC. Previous studies using the drug fucoxanthin have mainly focused on cell cycle arrest, induction of apoptosis, or inhibition of angiogenesis in GC. However, the effects of fucoxanthin on lymphangiogenesis and metastasis in GC have not been studied. METHODS: Cell counting kit 8 and transwell experiments were used to evaluate the inhibitory effect of fucoxanthin on cell proliferation, migration and invasion. HGC-27 and HLEC cells were co-cultured in a transwell chamber and the footpad metastasis model was established to evaluate lymphangiogenesis and lymph node metastasis. The possible regulatory targets of fucoxanthin in GC were analyzed using human tissue microarrays, bioinformatics analysis, and molecular docking. The regulatory pathway of fucoxanthin was verified using confocal laser microscopy, adenovirus transfection and western blotting. RESULTS: Tissue microarray and bioinformatics analyses showed that Ran was highly expressed in metastatic lymph nodes and has some predictive value for metastasis in GC. Molecular docking results revealed that fucoxanthin interacted with Met189 and Lys167 of Ran via hydrogen bonds. Mechanistically, fucoxanthin inhibits the nuclear transport of NF-κB by downregulating protein expression of Ran and importinß, thereby inhibiting VEGF-C secretion, and ultimately inhibiting tumor lymphangiogenesis and lymph node metastasis in vivo and in vitro. CONCLUSIONS: Fucoxanthin suppressed GC-induced lymphangiogenesis and metastasis in vitro and in vivo by regulating Ran expression via the importinß/NF-κB/VEGF-C nuclear transport signaling pathway. These novel findings provide the basis for the research and development of novel treatments using traditional Chinese medicine in treatment of lymph node metastasis, which has important theoretical significance and clinical value.


Subject(s)
Lymphangiogenesis , Stomach Neoplasms , Humans , Lymphatic Metastasis , NF-kappa B/metabolism , Vascular Endothelial Growth Factor C/metabolism , Stomach Neoplasms/drug therapy , Molecular Docking Simulation , Cell Line, Tumor
4.
Int J Mol Sci ; 24(9)2023 Apr 24.
Article in English | MEDLINE | ID: mdl-37175479

ABSTRACT

Secondary lymphedema is caused by lymphatic insufficiency (lymphatic drainage failure) following lymph node dissection during the surgical treatment or radiation therapy of breast or pelvic cancer. The clinical problems associated with lymphedema are reduced quality of life in terms of appearance and function, as well as the development of skin ulcers, recurrent pain, and infection. Currently, countermeasures against lymphedema are mainly physical therapy such as lymphatic massage, elastic stockings, and skin care, and there is no effective and fundamental treatment with a highly recommended grade. Therefore, there is a need for the development of a fundamental novel treatment for intractable lymphedema. Therapeutic lymphangiogenesis, which has been attracting attention in recent years, is a treatment concept that reconstructs the fragmented lymphatic network to recover lymphatic vessel function and is revolutionary to be a fundamental cure. This review focuses on the translational research of therapeutic lymphangiogenesis for lymphedema and outlines the current status and prospects in the development of therapeutic applications.


Subject(s)
Lymphangiogenesis , Lymphatic Vessels , Lymphedema , Humans , Lymph Node Excision/adverse effects , Lymphatic Vessels/pathology , Lymphedema/etiology , Lymphedema/therapy , Translational Research, Biomedical , Animals
5.
Phytomedicine ; 112: 154684, 2023 Apr.
Article in English | MEDLINE | ID: mdl-36738477

ABSTRACT

BACKGROUND: Macrophage-mediated inflammatory infiltration and pathological lymphangiogenesis around atherosclerotic plaques are newly highlighted treatment targets of atherosclerosis. Although the effect of Hydroxysafflor yellow A(HSYA) on atherosclerosis was clear, few studies focus on the regulation of HSYA on such mechanisms. PURPOSE: This study aimed to uncover the key site of HSYA on improving atherosclerosis by regulating macrophage-induced inflammation and lymphangiogenesis. STUDY DESIGN: This study was designed to explore the new mechanism of HSYA on alleviating atherosclerosis in vitro and in vivo. METHODS: We determined the expression of vascular endothelial growth factor C(VEGF-C) in Raw264.7 cells and high-fat diet fed ApoE knockout (ApoE-/-) mice. Raw264.7 cells were treated with HSYA under the stimulation of LPS and ox-LDL. HFD induced ApoE-/- mice were given different concentrations of HSYA-saline solution by tail vein injection and ATV-saline suspension by gavage. C57/B6j mice fed with chow diet were used for the control group. H&E, oil red O and immunofluorescence staining analysis were used for visualizing the pathological changes. The biological impact of HSYA was evaluated by body weight, lipid metabolism, inflammation levels, and corresponding function indexes of kidney and liver. RT-qPCR and western blot methods were conducted to determine the expression of the inflammation and lymphangiogenesis factors. Molecular docking and microscale thermophoresis analysis were used to verify the combination of HSYA and PI3K. RESULTS: In vivo, HSYA reduced the plaque formation, hepatic steatosis and inflammation-related lymphangiogenesis (IAL). It also changed the serum levels of inflammation (VEGF-C, TNF-α, IL-6, VCAM1, MCP1), lipid indexes (LDL, CHOL, TRIG) and relevant lymphangiogenesis (VEGF-C and LYVE-1) and inflammation (VCAM-1 and IL-6) signals in the aorta. In vitro, HSYA regulated Akt/mTOR and NF-κB activation by the inhibition of PI3K in macrophages. CONCLUSION: HSYA affects inflammation and inflammation-associated lymphangiogenesis via suppressing PI3K to affect AKT/mTOR and NF-B pathway activation in macrophages, showing a comprehensive protective effect on atherosclerosis.


Subject(s)
Atherosclerosis , NF-kappa B , Mice , Animals , NF-kappa B/metabolism , Vascular Endothelial Growth Factor C/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Interleukin-6/metabolism , Lymphangiogenesis , Molecular Docking Simulation , Mice, Knockout, ApoE , Atherosclerosis/drug therapy , Atherosclerosis/prevention & control , Atherosclerosis/metabolism , Macrophages , Inflammation/drug therapy , Inflammation/metabolism , TOR Serine-Threonine Kinases/metabolism , Apolipoproteins E
6.
Plast Reconstr Surg ; 151(4): 779-790, 2023 04 01.
Article in English | MEDLINE | ID: mdl-36729939

ABSTRACT

BACKGROUND: Decreased lymphangiogenesis contributes to impaired diabetic wound healing. Although negative-pressure wound therapy (NPWT) has been shown to be effective in the treatment of recalcitrant wounds, its impact on lymphangiogenesis remains to be elucidated. In this study, the authors investigate the mechanisms of lymphangiogenesis following NPWT treatment of diabetic murine wound healing. METHODS: Full-thickness dorsal skin wounds (1 × 1 cm 2 ) were excised on 30 db/db mice. The mice were either treated with occlusive covering (control group, n = 15), or received a 7-day treatment of continuous NPWT at -125 mmHg (NPWT group, n = 15). The wounds were photographed on days 0, 7, 10, 14, 21, and 28. Wound tissue was harvested on days 10, 14, 21, and 28 for quantitative analysis. Functional analysis of lymphatic drainage was performed on days 14 and 28 with Evans blue dye tracing. RESULTS: Lymphatic density and diameter, as visualized through podoplanin probing, was significantly higher in the NPWT group compared to the control group ( P < 0.001). NPWT up-regulated the expression of lymphatic vessel endothelial hyaluronan receptor 1 (LYVE-1) at the protein level ( P = 0.04), and significant differences were noted in lymphatic density as assessed by LYVE-1 staining ( P = 0.001). Leukocyte infiltration was significantly higher in the NPWT group ( P = 0.01). A higher speed of wound closure ( P < 0.0001) and greater wound bed thickness ( P < 0.0001) were noted in the NPWT group compared to the control group. CONCLUSIONS: NPWT increased the lymphatic vessel density and diameter with LYVE-1 up-regulation. NPWT therefore plays a positive role in lymphangiogenesis in diabetic wound healing. CLINICAL RELEVANCE STATEMENT: The authors' study investigates the association of NPWT and lymphatics and underlines the importance of a more in-depth investigation of the role of lymphatic vessels in wound healing.


Subject(s)
Diabetes Mellitus , Negative-Pressure Wound Therapy , Soft Tissue Injuries , Mice , Animals , Lymphangiogenesis , Wound Healing , Soft Tissue Injuries/therapy
7.
Adv Wound Care (New Rochelle) ; 12(9): 483-497, 2023 Sep.
Article in English | MEDLINE | ID: mdl-36424821

ABSTRACT

Objective: Despite the significant function of lymphatics in wound healing, and frequent clinical use of Negative Pressure Wound Therapy (NPWT), the effect of mechanical force application on lymphangiogenesis remains to be elucidated. We utilize a murine incisional wound healing model to assess the mechanisms of lymphangiogenesis following NPWT. Approach: Dorsal incisional skin wounds were created on diabetic mice (genetically obese leptin receptor-deficient mice [db/db]; n = 30) and covered with an occlusive dressing (Control, n = 15) or NPWT (-125 mmHg, continuous, 24 h for 7 days; NPWT, n = 15). The wounds were macroscopically assessed for 28 days. Tissue was harvested on day 10 for analysis. Qualitative functional analysis of lymphatic drainage was performed on day 28 using Evans Blue staining (n = 2). Results: NPWT increased lymphatic vessel density (40 ± 20 vs. 12 ± 6 podoplanin [PDPN]+ and 25 ± 9 vs. 14 ± 8 lymphatic vessel endothelial receptor 1 [LYVE-1]+) and vessel diameter (28 ± 9 vs. 12 ± 2 µm). Western blotting verified the upregulation of LYVE-1 with NPWT. Leukocyte presence was higher with NPWT (22% ± 3.7% vs. 9.1% ± 4.1% lymphocyte common antigen [CD45]+) and the leukocytes were predominately B cells clustered within vessels (8.8% ± 2.5% vs. 18% ± 3.6% B-lymphocyte antigen CD20 [CD20]+). Macrophage presence was lower in the NPWT group. Lymphatic drainage was increased in the NPWT group, which exhibited greater Evans Blue positivity. Innovation: The lymphangiogenic effects take place independent of macrophage infiltration, appearing to correlate with B cell presence. Conclusion: NPWT promotes lymphangiogenesis in incisional wounds, significantly increasing the lymph vessel density and diameter. This study highlights the potential of NPWT to stimulate lymphatic drainage and wound healing of surgical incisions.


Subject(s)
Diabetes Mellitus, Experimental , Negative-Pressure Wound Therapy , Animals , Mice , Lymphangiogenesis , Evans Blue , Wound Healing/physiology
8.
Autophagy ; 19(4): 1351-1353, 2023 04.
Article in English | MEDLINE | ID: mdl-36026459

ABSTRACT

Lymphatic endothelial cells (LECs) exploit fatty acid oxidation (FAO) to grow and to maintain lymphatic vessel identity through the epigenetic regulation of the essential transcription factor PROX1. In our recent study, we found that LEC-specific loss of ATG5 prevents injury-induced lymphangiogenesis in vivo. Inadequate degradation of lipid droplets (LDs) caused by genetic ablation of ATG5 in LECs disturbs mitochondrial fitness, and reduces mitochondrial FAO and acetyl-CoA levels, ultimately affecting PROX1-mediated epigenetic regulation of CPT1A and key lymphatic markers, most importantly FLT4/VEGFR3. Supplementing the fatty acid precursor acetate rescues defective inflammation-driven lymphangiogenesis in LEC-specific atg5 knockout mice. Thus, efficient macroautophagy/autophagy-mediated LD breakdown is critical to maintain mitochondrial metabolism and acetyl-CoA levels, which sustain a PROX1-mediated lymphatic gene program required for LEC identity and inflammation-driven lymphangiogenesis.


Subject(s)
Endothelial Cells , Lymphangiogenesis , Mice , Animals , Endothelial Cells/metabolism , Epigenesis, Genetic , Acetyl Coenzyme A/metabolism , Homeodomain Proteins/genetics , Tumor Suppressor Proteins/metabolism , Autophagy , Inflammation/metabolism , Mice, Knockout , Fatty Acids/metabolism
9.
Biomed Pharmacother ; 154: 113630, 2022 Oct.
Article in English | MEDLINE | ID: mdl-36058147

ABSTRACT

Gastric cancer (GC) is one of the most common gastrointestinal malignancies in the world. Growing evidence emphasizes the critical role of long non-coding RNA (lncRNA) in GC tumorigenesis. The aim of the research was to elucidate the effect and mechanism of Babao Dan (BBD) on lymphangiogenesis of GC in vitro and in vivo via lncRNA-ANRIL/VEGF-C/VEGFR-3 signaling axis. The present study investigated BBD significantly decreased the expression of lncRNA-ANRIL and VEGF-C in GC cells (AGS, BGC823, and MGC80-3) by using real-time quantitative polymerasechain reaction (RT-qPCR) and the secretion and expression of VEGF-C by (enzyme linked immunosorbent assay) ELISA and western blot (WB). BBD significantly inhibited the tumor xenograft of GC growth and the expression of lncRNA-ANRIL, VEGF-C, VEGFR-3 and LYVE-1 in vivo. BBD reduced serum VEGF-C level. In vitro, BBD inhibited the tube formation and decreased the cell viability, proliferation and migration of HLECs by using tube formation, MTT, Hoechst and Transwell assays. In addition, WB assay found that BBD decreased the expression levels of VEGF-C, VEGFR-3, matrix metallopeptidase 2 (MMP-2) and matrix metallopeptidase 9 (MMP-9), and RT-qPCR assay found that the mRNA expression levels of lncRNA-ANRIL, VEGF-C, VEGFR-3, MMP-2, MMP-9, CDK4, Cyclin D1, and Bcl-2 were down-regulated, and the expression of p21 and Bax were increased. Taken together, these results demonstrated that BBD inhibited lymphangiogenesis of GC in vitro and in vivo via the lncRNA-ANRIL/VEGF-C/VEGFR-3 signaling axis.


Subject(s)
RNA, Long Noncoding , Stomach Neoplasms , Cell Line, Tumor , Drugs, Chinese Herbal , Humans , Lymphangiogenesis/genetics , Matrix Metalloproteinase 2 , Matrix Metalloproteinase 9 , RNA, Long Noncoding/genetics , RNA, Long Noncoding/pharmacology , Stomach Neoplasms/drug therapy , Stomach Neoplasms/genetics , Stomach Neoplasms/metabolism , Vascular Endothelial Growth Factor C/genetics , Vascular Endothelial Growth Factor C/metabolism , Vascular Endothelial Growth Factor Receptor-3/genetics , Vascular Endothelial Growth Factor Receptor-3/metabolism
10.
J Food Biochem ; 46(9): e14233, 2022 09.
Article in English | MEDLINE | ID: mdl-35567300

ABSTRACT

Administration of Piper retrofractum extract (PRE) has been reported to alleviate edema, but the mechanism underlying this effect is unknown. Promotion of lymphangiogenesis is known to improve lymphedema, but the effect of PRE on lymphangiogenesis remains unclear. In the present study, we investigated whether PRE and specifically, piperine, the main component of PRE, can induce lymphangiogenesis. Treatments with PRE and piperine significantly promoted the proliferation, migration, and tube formation in human dermal lymphatic microvascular endothelial cells (HDLECs) but had no effect on the expression of lymphangiogenic factors. Furthermore, PRE and piperine significantly promoted the phosphorylation of the AKT and ERK proteins in HDLECs, and pretreatment with AKT and ERK inhibitors significantly attenuated the PRE- and piperine-induced lymphangiogenesis. These results indicate that PRE and piperine promote lymphangiogenesis via an AKT- and ERK-dependent mechanism. PRACTICAL APPLICATIONS: The lymphatic system plays various roles such as maintaining tissue fluid homeostasis, immune defense, and metabolism. Disruption of the lymphatic system results in insufficient fluid drainage, which causes edema. Currently, there are no effective treatments for lymphedema; therefore, the development of novel treatment strategies is desirable. In this study, we showed that PRE and its main component piperine promote lymphangiogenesis in lymphatic endothelial cells. Therefore, PRE has the potential to be used as a novel functional food for relieving lymphedema.


Subject(s)
Lymphedema , Piper , Alkaloids , Benzodioxoles , Endothelial Cells/metabolism , Humans , Lymphangiogenesis , Lymphedema/drug therapy , Lymphedema/metabolism , Piperidines , Plant Extracts/metabolism , Plant Extracts/pharmacology , Polyunsaturated Alkamides , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism
11.
Nat Commun ; 13(1): 2760, 2022 05 19.
Article in English | MEDLINE | ID: mdl-35589749

ABSTRACT

Autophagy has vasculoprotective roles, but whether and how it regulates lymphatic endothelial cells (LEC) homeostasis and lymphangiogenesis is unknown. Here, we show that genetic deficiency of autophagy in LEC impairs responses to VEGF-C and injury-driven corneal lymphangiogenesis. Autophagy loss in LEC compromises the expression of main effectors of LEC identity, like VEGFR3, affects mitochondrial dynamics and causes an accumulation of lipid droplets (LDs) in vitro and in vivo. When lipophagy is impaired, mitochondrial ATP production, fatty acid oxidation, acetyl-CoA/CoA ratio and expression of lymphangiogenic PROX1 target genes are dwindled. Enforcing mitochondria fusion by silencing dynamin-related-protein 1 (DRP1) in autophagy-deficient LEC fails to restore LDs turnover and lymphatic gene expression, whereas supplementing the fatty acid precursor acetate rescues VEGFR3 levels and signaling, and lymphangiogenesis in LEC-Atg5-/- mice. Our findings reveal that lipophagy in LEC by supporting FAO, preserves a mitochondrial-PROX1 gene expression circuit that safeguards LEC responsiveness to lymphangiogenic mediators and lymphangiogenesis.


Subject(s)
Lymphangiogenesis , Lymphatic Vessels , Animals , Autophagy/genetics , Endothelial Cells/metabolism , Fatty Acids/metabolism , Lipid Droplets/metabolism , Lymphangiogenesis/genetics , Lymphatic Vessels/metabolism , Mice , Mitochondria , Transcription Factors/metabolism
12.
Biomed Pharmacother ; 147: 112664, 2022 Mar.
Article in English | MEDLINE | ID: mdl-35131655

ABSTRACT

The lymphatic vascular system is crucial for maintaining tissue fluid homeostasis and immune surveillance. Promoting lymphatic function represents a new strategy to treat several diseases including lymphedema, chronic inflammation and impaired wound healing. By screening a plant extract library, a petroleum ether extract from the aerial parts of Eupatorium perfoliatum (E. perfoliatum) was found to possess lymphangiogenic properties. With the aid of HPLC activity profiling the active compound was identified as pheophorbide a. Both plant extract and pheophorbide a induced the sprouting and tube formation of human primary lymphatic endothelial cells (LECs). The proliferation of the LECs was increased upon treatment with pheophorbide a but not the E. perfoliatum extract. Treatment with the MEK1/2 inhibitor U0126 reduced the LEC sprouting activity, indicating a potential mechanism of action. These studies suggest that pheophorbide a could represent novel natural therapeutic agent to treat human lymphatic vascular insufficiencies.


Subject(s)
Chlorophyll/analogs & derivatives , Endothelial Cells/drug effects , Eupatorium , Lymphangiogenesis/drug effects , Plant Extracts/pharmacology , Butadienes/pharmacology , Cell Line , Chlorophyll/pharmacology , Humans , Lymphatic Vessels/drug effects , MAP Kinase Kinase 1/antagonists & inhibitors , MAP Kinase Kinase 2/antagonists & inhibitors , Nitriles/pharmacology
13.
Phytother Res ; 34(2): 359-367, 2020 Feb.
Article in English | MEDLINE | ID: mdl-31680342

ABSTRACT

Tumor angiogenesis and lymphangiogenesis are important processes in tumor progression and metastasis. The inhibitory effects of 3-O-acetyloleanolic acid (3AOA), a pentacyclic triterpenoid compound isolated from Vigna sinensis K., on tumor-induced angiogenesis and lymphangiogenesis in vitro and in vivo were studied. Angiopoietin-1 is an important angiogenic and lymphangiogenic factor secreted from colon carcinoma CT-26 cells under hypoxia conditions. 3AOA inhibited proliferation, migration, and tube formation of angiopoietin-1-treated human umbilical vein endothelial cells (HUVEC) and human lymphatic microvascular endothelial cells (HLMEC). 3AOA reduced angiogenesis and lymphangiogenesis in angiopoietin-1-stimulated Matrigel plugs. Also, 3AOA inhibited tumor growth and tumor-induced angiogenesis and lymphangiogenesis in an angiopoietin-1-induced CT-26 allograft colon carcinoma animal model. 3AOA inhibited activation of the angiopoietin-1 receptor Tie-2 and activation of the downstream signaling factors FAK, AKT, and ERK1/2 that are involved in the angiopoietin-1/Tie-2-signaling pathway. Thus, 3AOA has an inhibitory effect on angiogenesis and lymphangiogenesis induced by angiopoietin-1 both in vitro and in vivo, and the inhibitory effect of 3AOA is probably due to suppression of angiopoietin-1/Tie-2 signaling in HUVEC and HLMEC.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Angiopoietin-1/metabolism , Lymphangiogenesis/drug effects , Neovascularization, Pathologic/drug therapy , Receptor, TIE-2/metabolism , Triterpenes/pharmacology , Animals , Apoptosis/drug effects , Cell Line, Tumor , Female , Human Umbilical Vein Endothelial Cells/drug effects , Humans , Mice , Mice, Inbred BALB C , Neoplasms, Experimental/drug therapy , Signal Transduction/drug effects
14.
Oncol Rep ; 42(3): 1225-1236, 2019 Sep.
Article in English | MEDLINE | ID: mdl-31322263

ABSTRACT

Colorectal cancer (CRC) is one of the most commonly diagnosed malignancies worldwide. For patients diagnosed with the presence of metastatic disease, surgery is not suitable for the majority of them. Lymphangiogenesis is a key factor during cancer metastasis and is regulated by vascular endothelial growth factor C (VEGF­C). Hedyotis diffusa Willd. (HDW) is a Chinese herb of the Rubiaceae family that reportedly inhibits tumor metastasis. However, its underlying anticancer mechanisms have not yet been elucidated. In the present study, we investigated the effects of an ethanol extract of HDW (EEHDW) on the migration capacity by wound healing and Transwell assays, and the effect on the VEGF­C expression in different CRC cell lines by western blot analysis and ELISA assays. A model of VEGF­C­stimulated human lymphatic endothelial cells (HLECs) was constructed. It was found that EEHDW suppressed lymphangiogenesis via the mediation of multiple pathways, which attenuated the migration of cells and their tube formation abilities. Multiple signaling pathways were found to be involved in the VEGF­C­mediated lymphangiogenesis. After EEHDW treatment in VEGF­C­stimulated HLECs, EEHDW was found to downregulate the expression levels of multiple signaling pathways. Taken together, these results indicate that EEHDW possesses significant anti­metastatic activities. Moreover, the suppressive effect of EEHDW on lymphangiogenesis, particularly via downregulation of VEGF­C, partly explains the potential molecular mechanism underlying the inhibitory effect of EEHDW on CRC metastasis.


Subject(s)
Colorectal Neoplasms/drug therapy , Gene Expression Regulation, Neoplastic/drug effects , Hedyotis/chemistry , Lymphangiogenesis/drug effects , Plant Extracts/pharmacology , Signal Transduction/drug effects , Vascular Endothelial Growth Factor C/metabolism , Apoptosis , Cell Movement , Cell Proliferation , Colorectal Neoplasms/blood supply , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , Humans , Tumor Cells, Cultured
15.
Arthritis Rheumatol ; 71(2): 244-257, 2019 02.
Article in English | MEDLINE | ID: mdl-30144298

ABSTRACT

OBJECTIVE: To investigate the roles of the synovial lymphatic system in the severity and progression of joint tissue damage and functional responses of synovial lymphatic endothelial cells (LECs) to macrophage subsets, and to evaluate the therapeutic potential of the proteasome inhibitor bortezomib (BTZ) in a mouse model of experimental posttraumatic osteoarthritis (OA). METHODS: C57BL/6J wild-type mice received a meniscal ligamentous injury to induce posttraumatic knee OA. Lymphangiogenesis was blocked by a vascular endothelial growth factor receptor 3 (VEGFR-3) neutralizing antibody. Synovial lymphatic drainage was examined by near-infrared imaging. Joint damage was assessed by histology. RNA-sequencing and pathway analyses were applied to synovial LECs. Macrophage subsets in the mouse synovium were identified by flow cytometry and immunofluorescence staining. M1 and M2 macrophages were induced from mouse bone marrow cells, and their effects on LECs were examined in cocultures in the presence or absence of BTZ. The effects of BTZ on joint damage, LEC inflammation, and synovial lymphatic drainage were examined. RESULTS: Injection of a VEGFR-3 neutralizing antibody into the joints of mice with posttraumatic knee OA reduced synovial lymphatic drainage and accelerated joint tissue damage. Synovial LECs from the mouse OA joints had dysregulated inflammatory pathways and expressed high levels of inflammatory genes. The number of M1 macrophages was increased in the knee joints of mice with posttraumatic OA, thereby promoting the expression of inflammatory genes by LECs; this effect was blocked by BTZ. Treatment with BTZ decreased cartilage loss, reduced the expression of inflammatory genes by LECs, and improved lymphatic drainage in the knee joints of mice with posttraumatic OA. CONCLUSION: Experimental posttraumatic knee OA is associated with decreased synovial lymphatic drainage, increased numbers of M1 macrophages, and enhanced inflammatory gene expression by LECs, all of which was improved by treatment with BTZ. Intraarticular administration of BTZ may represent a new therapy for the restoration of synovial lymphatic function in subjects with posttraumatic knee OA.


Subject(s)
Bortezomib/pharmacology , Endothelial Cells/drug effects , Lymphatic Vessels/drug effects , Macrophages/drug effects , Osteoarthritis, Knee/immunology , Proteasome Inhibitors/pharmacology , Synovial Membrane/drug effects , Animals , Antibodies, Neutralizing/pharmacology , Bortezomib/therapeutic use , Coculture Techniques , Disease Progression , Inflammation , Knee Injuries/complications , Lymphangiogenesis/drug effects , Lymphatic Vessels/immunology , Macrophages/immunology , Mice , Osteoarthritis, Knee/drug therapy , Osteoarthritis, Knee/etiology , Proteasome Inhibitors/therapeutic use , Spectroscopy, Near-Infrared , Vascular Endothelial Growth Factor Receptor-3/antagonists & inhibitors , Vascular Endothelial Growth Factor Receptor-3/immunology
16.
Biol Pharm Bull ; 41(11): 1659-1666, 2018.
Article in English | MEDLINE | ID: mdl-30381665

ABSTRACT

Lymphangiogenesis, the formation of lymphatic vessels from preexisting ones, promotes cancer growth and metastasis. Finding natural compounds with anti-lymphangiogenic activity will be useful for preventive treatment of lymphatic metastasis. Shikonin, an ingredient of a traditional Japanese and Chinese medicinal herb Lithospermum erythrorhizon, has been widely used in several pharmaceutical and cosmetic preparations, as well as in food colorants. Shikonin has been reported to inhibit lymphangiogenesis in vitro, but the mechanism of inhibition has not been determined. The aim of this study is to investigate the mechanism of anti-lymphangiogenesis of shikonin in primary human lymphatic endothelial cells (HMVEC-dLy). Shikonin, at non-toxic concentrations, significantly inhibited cord formation ability of lymphatic endothelial cells in a dose- and time-dependent manner. Western blotting analysis showed that shikonin decreased nuclear factor-kappaB (NF-κB) activation, as indicated by phosphorylation and nuclear translocation of NF-κB p65, and also reduced both mRNA and protein levels of hypoxia-inducible factor-1 (HIF-1)α. Use of an NF-κB inhibitor (BAY 11-7085) and HIF-1α small interfering RNA (siRNA) transfection revealed that NF-κB activation was upstream of HIF-1α expression, which controls cord formation by HMVEC-dLy. In addition, the reduction of vascular endothelial growth factor C (VEGF-C) and vascular endothelial growth factor receptor-3 (VEGFR-3) mRNA levels were also found in HMVEC-dLy that treated with shikonin. In conclusion, shikonin inhibits lymphangiogenesis in vitro by interfering the NF-κB/HIF-1α pathway and involves in suppression of VEGF-C and VEGFR-3 mRNA expression.


Subject(s)
Drugs, Chinese Herbal/pharmacology , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Lithospermum/chemistry , Lymphangiogenesis/drug effects , NF-kappa B/metabolism , Naphthoquinones/pharmacology , Drugs, Chinese Herbal/therapeutic use , Endothelial Cells/drug effects , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Lymphatic Metastasis/prevention & control , Naphthoquinones/therapeutic use , Phytotherapy , RNA, Messenger/metabolism , Vascular Endothelial Growth Factor C/genetics , Vascular Endothelial Growth Factor C/metabolism , Vascular Endothelial Growth Factor Receptor-3/genetics , Vascular Endothelial Growth Factor Receptor-3/metabolism
17.
Microcirculation ; 25(8): e12502, 2018 11.
Article in English | MEDLINE | ID: mdl-30178505

ABSTRACT

OBJECTIVE: Motivated by observations of mesenteries harvested from mice treated with tamoxifen dissolved in oil for inducible gene mutation studies, the objective of this study was to demonstrate that microvascular growth can be induced in the avascular mouse mesentery tissue. METHODS: C57BL/6 mice were administered an IP injection for five consecutive days of: saline, sunflower oil, tamoxifen dissolved in sunflower oil, corn oil, or peanut oil. RESULTS: Twenty-one days post-injection, zero tissues from saline group contained branching microvascular networks. In contrast, all tissues from the three oils and tamoxifen groups contained vascular networks with arterioles, venules, and capillaries. Smooth muscle cells and pericytes were present in their expected locations and wrapping morphologies. Significant increases in vascularized tissue area and vascular density were observed when compared to saline group, but sunflower oil and tamoxifen group were not significantly different. Vascularized tissues also contained LYVE-1-positive and Prox1-positive lymphatic networks, indicating that lymphangiogenesis was stimulated. When comparing the different oils, vascularized tissue area and vascular density of sunflower oil were significantly higher than corn and peanut oils. CONCLUSIONS: These results provide novel evidence supporting that induction of microvascular network growth into the normally avascular mouse mesentery is possible.


Subject(s)
Mesentery/blood supply , Microvessels/drug effects , Plant Oils/pharmacology , Tamoxifen/pharmacology , Animals , Lymphangiogenesis , Mesentery/pathology , Mice , Mice, Inbred C57BL , Microvessels/growth & development , Neovascularization, Physiologic/drug effects
18.
J Nat Med ; 72(2): 481-492, 2018 Mar.
Article in English | MEDLINE | ID: mdl-29350326

ABSTRACT

The purpose of this study was to investigate the effects and mechanisms of oxyresveratrol (Oxyres) on hepatocellular carcinoma (HCC) in vitro and in vivo. The MTT and Transwell assays were performed to investigate the effects of Oxyres on cell proliferation and migration of two HCC cell lines, QGY-7701 and SMMC-7721 cells. H22 cells were subcutaneously injected into hind foot pads of 70 male mice to establish a lymph node metastasis model. These mice were randomly divided into seven groups as follows, control group, HCC group, Oxyres 20 mg/kg group, Oxyres 40 mg/kg group, Oxyres 60 mg/kg group, Resveratrol (Res) group, and Adriamycin (ADM) group. Oxyres, Res, and ADM were intraperitoneally injected daily for consecutive 21 days. Tumors and popliteal lymph node were isolated and embedded for histology analysis. Expressions of CD31 and vascular endothelial growth factor receptor-3 (VEGFR3) in tumors were detected by immunohistocehmistry. Expressions of vascular endothelial growth factor C (VEGF-C) were measured by Western blot. Oxyres significantly inhibited the proliferation and migration of QGY-7701 and SMMC-7721 cells. Oxyres significantly inhibited tumor growth (p < 0.001) and metastasis to sentinel lymph nodes (70%) in a dose-dependent manner. Oxyres showed a similar inhibition rate as Res. Oxyres also significantly decreased micro-blood vessel density and micro-lymphatic vessel density in tumors (p < 0.05). Expressions of CD31, VEGFR3, and VEGF-C of tumors were also inhibited by Oxyres (p < 0.05). Oxyres exerts anti-tumor effects against HCC through inhibiting both angiogenesis and lymph node metastasis, which suggests Oxyres be a potential therapeutic agent.


Subject(s)
Angiogenesis Inhibitors/therapeutic use , Carcinoma, Hepatocellular/drug therapy , Liver Neoplasms/drug therapy , Lymphangiogenesis/drug effects , Lymphatic Metastasis/prevention & control , Plant Extracts/therapeutic use , Stilbenes/therapeutic use , Angiogenesis Inhibitors/pharmacology , Animals , Carcinoma, Hepatocellular/pathology , Cell Culture Techniques , Humans , Liver Neoplasms/pathology , Male , Mice , Plant Extracts/pharmacology , Stilbenes/pharmacology
19.
Eur J Cancer ; 83: 19-27, 2017 09.
Article in English | MEDLINE | ID: mdl-28709135

ABSTRACT

Photodynamic therapy (PDT) has been shown to destroy tumour-associated lymphatic vessels. Therefore, we sought to investigate the functional outcomes of PDT-mediated damage to the lymphatic vessels. We observed that PDT with verteporfin, completely but transiently, blocks the functional lymphatic drainage in the orthotopic mammary tumour models. Sustained inhibition of lymphatic vessels regeneration induced by lenalidomide or the soluble form of vascular endothelial growth factor receptor 3 (sVEGFR3) that neutralises lymphangiogenic vascular endothelial growth factor C (VEGF-C), significantly impaired antitumour efficacy of PDT. Antilymphangiogenic compounds also significantly inhibited the ability of intratumourally inoculated dendritic cells (DCs) to translocate to local lymph nodes and diminished the number of tumour-infiltrating interferon-γ-secreting or tumour antigen-specific CD8+ T cells. Lenalidomide also abrogated antitumour effects of the combination immunotherapy with PDT and anti-programmed death-ligand 1 (PD-L1) antibodies. Altogether, these findings indicate that PDT-mediated damage to the lymphatic vessels negatively affects development of antitumour immunity, and that drugs that impair lymphatic vessel regeneration might not be suitable for the use in combination with PDT.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Antineoplastic Agents/pharmacology , Breast Neoplasms/drug therapy , Cell Cycle Checkpoints/drug effects , Lymphangiogenesis/drug effects , Photochemotherapy , Porphyrins/metabolism , Porphyrins/pharmacology , Thalidomide/analogs & derivatives , Animals , Disease Models, Animal , Female , Lenalidomide , Lymphangiogenesis/radiation effects , Lymphatic Vessels/drug effects , Lymphatic Vessels/pathology , Mice , Mice, Inbred C57BL , Photosensitizing Agents/pharmacology , Thalidomide/pharmacology , Vascular Endothelial Growth Factor Receptor-3/pharmacology , Verteporfin
20.
Oncotarget ; 8(19): 32027-32042, 2017 May 09.
Article in English | MEDLINE | ID: mdl-28410190

ABSTRACT

Previously, we reported that high-fat-diet (HFD)-induced obesity stimulates melanoma progression in the B16F10 allograft model. In this study, we examined whether oleuropein (OL), the most abundant phenolic compound in olives, inhibits HFD-induced melanoma progression. Four-week-old male C57BL/6N mice were fed a HFD-diet with or without OL. After 16 weeks of feeding, B16F10-luc cells were subcutaneously injected and the primary tumor was resected 3 weeks later. OL suppressed HFD-induced solid tumor growth. In the tumor tissues, OL reduced HFD-induced expression of angiogenesis (CD31, VE-cadherin, VEGF-A, and VEGFR2), lymphangiogenesis (LYVE-1, VEGF-C, VEGF-D, and VEGFR3), and hypoxia (HIF-1α and GLUT-1) markers as well as HFD-induced increases in lipid vacuoles and M2 macrophages (MΦs). All animals were euthanized 2.5 weeks after tumor resection. OL suppressed HFD-induced increases in lymph node (LN) metastasis; expression of VEGF-A, VEGF-C, and VEGF-D in the LN; and M2-MΦs and the size of adipocytes in adipose tissues surrounding LNs. Co-culture results revealed that the crosstalk between B16F10s, M2-MΦs, and differentiated 3T3-L1 cells under hypoxic conditions increased the secretion of VEGF-A and -D, which stimulated tube formation and migration of endothelial cells (HUVECs) and lymphatic endothelial cells (LEC), respectively. Additionally, OL directly inhibited the differentiation of 3T3-L1 preadipocytes and tube formation by HUVECs and LECs. The overall results indicated that dietary OL inhibits lipid and M2-MΦ accumulation in HFD-fed mice, which contributes to decreases in VEGF secretion, thereby leading to inhibition of angiogenesis and lymphangiogenesis.


Subject(s)
Angiogenesis Inhibitors/administration & dosage , Dietary Supplements , Iridoids/administration & dosage , Lymphangiogenesis/drug effects , Melanoma, Experimental/pathology , Neovascularization, Pathologic , Adipose Tissue/drug effects , Adipose Tissue/metabolism , Allografts , Animals , Apoptosis , Cell Proliferation/drug effects , Diet, High-Fat , Hypoxia/metabolism , Iridoid Glucosides , Lipid Metabolism , Lymphatic Metastasis , Macrophages/drug effects , Macrophages/metabolism , Melanoma, Experimental/drug therapy , Mice , Neovascularization, Pathologic/drug therapy , Tumor Burden , Vascular Endothelial Growth Factors/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL