Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 57
Filter
1.
Microvasc Res ; 140: 104305, 2022 03.
Article in English | MEDLINE | ID: mdl-34958805

ABSTRACT

Angiogenesis is critical for solid tumor growth beyond its minimal size. Previously, we reported that Down Syndrome Candidate Region 1 isoform 1L (DSCR1-1L) was one of the most up-regulated genes in endothelial cells induced by VEGF and histamine, and regulated endothelial cell proliferation, migration and angiogenesis. However, it was not known whether DSCR1-1L played a role in tumor growth. In this study, we found that DSCR1-1L shRNAs significantly inhibited the growth of transplanted melanoma in mice and its associated tumoral angiogenesis. In the gain of function assay, overexpression of DSCR1-1L cDNA in mouse endothelium is sufficient to significantly increase the tumor initiation induced by carcinogen, the growth of xenografted tumor, and the tumor metastasis in our endothelially-expressed DSCR1-1L transgenic mice, in which angiogenesis was induced. It was the first time to find that DSCR1-1L was also expressed in various tumor cells. DSCR1-1L shRNAs inhibited, but overexpression of DSCR1-1L cDNA increased, the tumor cell proliferation and migration. Most recently, we reported that DSCR1-1L modulated angiogenesis by down-regulation of VE-cadherin expression. Here, we found that DSCR1-1L down-regulated the expression of E-cadherin. Hence, DSCR1-1L is an excellent therapeutic target for cancers by regulation of both the endothelial and tumor cells through down-regulating (V)E-cadherin. DSCR1-1L shRNAs have the potential to be developed for clinical application.


Subject(s)
Calcium-Binding Proteins/metabolism , DNA-Binding Proteins/metabolism , Endothelial Cells/metabolism , Melanoma/blood supply , Melanoma/metabolism , Muscle Proteins/metabolism , Neovascularization, Pathologic , Skin Neoplasms/blood supply , Skin Neoplasms/metabolism , Animals , Antigens, CD/genetics , Antigens, CD/metabolism , Cadherins/genetics , Cadherins/metabolism , Calcium-Binding Proteins/genetics , Cell Line, Tumor , Cell Movement , Cell Proliferation , DNA-Binding Proteins/genetics , Endothelial Cells/pathology , Female , Gene Expression Regulation, Neoplastic , Humans , Male , Melanoma/genetics , Melanoma/pathology , Mice, Nude , Muscle Proteins/genetics , Neoplasm Invasiveness , Protein Isoforms , Signal Transduction , Skin Neoplasms/genetics , Skin Neoplasms/pathology , Tumor Burden
2.
Microvasc Res ; 138: 104209, 2021 11.
Article in English | MEDLINE | ID: mdl-34146582

ABSTRACT

Angiogenesis is critical for many diseases. Previously, we reported that Down Syndrome Candidate Region 1 isoform 1L (DSCR1-1L) was one of the most up-regulated genes in endothelial cells induced by VEGF and histamine, and regulated endothelial cell proliferation and Matrigel angiogenesis in mice. However, it was not known whether DSCR1-1L regulated angiogenesis in vivo and what was the molecular mechanism underlying it. In this study, gene knockdown and overexpression models were established to study the role of DSCR1-1L in angiogenesis in vivo. Further, the downstream regulatory target of DSCR1-1L was explored with molecular biological methods in vascular endothelial cells. We found that DSCR1-1L shRNAs significantly inhibited angiogenesis induced by VEGF in mice (p < 0.0001). In the gain-of-function assay, overexpression of DSCR1-1L cDNA in mouse endothelium of EC-FH-DSCR1-1L transgenic mice was sufficient to induce angiogenesis significantly (p < 0.01). DSCR1-1L regulated angiogenesis in the early stage by down-regulation of the VE-cadherin expression through targeting its transcription, but not mRNA stability. Three DSCR1-1L-targeted DNA elements in the VE-cadherin promoter were identified by promoter reporter assays, among which, a novel specific transcriptional complex was found. The DNA sequence (CTTCTG) in the VE-cadherin promoter was identified to directly interact with proteins by Electrophoresis Mobility Shift Assays and DNase I footprint assay. Hence, DSCR1-1L is an excellent therapeutic target for angiogenic diseases through down-regulating the formation of a novel transcriptional complex on the VE-cadherin promoter. DSCR1-1L shRNAs and cDNA have the potential to be developed for clinical application. Our results also contribute significantly to the field of mechanistic studies.


Subject(s)
Antigens, CD/metabolism , Cadherins/metabolism , Human Umbilical Vein Endothelial Cells/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Melanoma, Experimental/blood supply , Muscle Proteins/metabolism , Neovascularization, Pathologic , Neovascularization, Physiologic , Promoter Regions, Genetic , Animals , Antigens, CD/genetics , Cadherins/genetics , Cell Line, Tumor , Cell Movement , Cell Proliferation , Down-Regulation , Female , Humans , Intracellular Signaling Peptides and Proteins/genetics , Male , Melanoma, Experimental/genetics , Melanoma, Experimental/metabolism , Mice, Nude , Mice, Transgenic , Muscle Proteins/genetics , Signal Transduction
3.
Microvasc Res ; 129: 103955, 2020 05.
Article in English | MEDLINE | ID: mdl-31733305

ABSTRACT

Angiogenesis is a hallmark of many diseases. Previously, we found that Down Syndrome Candidate Region 1 Isoform 1L (DSCR1-1L) was expressed in human tumor vessels, but was not detectable in normal tissues, and played important roles in angiogenesis induced by vascular endothelial growth factor (VEGF-A165). The expressions of DSCR1-1L mRNA and protein induced by VEGF-A165 were regulated via the direct interaction of transcription enhancer factor 3 (TEF3) with DSCR1-1L promoter. However, the function and the regulation of DSCR1-1L in angiogenesis had not been completely understood. In this study, we found that the expressions of DSCR1-1L mRNA and proteins were upregulated by other angiogenic factors, including VEGF-A121, VEGF-E, histamine, PAF, the endothelial cell (EC) growth medium, and the conditional medium obtained from cancer cells, but not by PlGF, bFGF, PDGF, and serotonin. The EC proliferation, migration and elongation induced by histamine and EC growth medium were inhibited by knocking down the mRNA and protein expressions of DSCR1-1L and TEF3. The TEF3 activation was regulated by its interaction with YAP1, and translocation from cytosol to nuclei, but not by increase of protein expression, after the stimulation of VEGF, histamine and EC growth medium. YAP1 regulated the protein expression of DSCR1-1L, the proliferation, migration and elongation of ECs induced by VEGF, histamine and EC growth medium. Taken together, this study identified a novel axis of YAP1, TEF3 and DSCR1-1L that was a common signaling pathway downstream of several angiogenic factors to regulate angiogenesis, suggesting that this pathway is an excellent therapeutic target for angiogenic diseases and cancers. Our results contribute significantly to the field of mechanistic studies.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Angiogenesis Inducing Agents/pharmacology , DNA-Binding Proteins/metabolism , Human Umbilical Vein Endothelial Cells/drug effects , Muscle Proteins/metabolism , Neovascularization, Physiologic/drug effects , Transcription Factors/metabolism , Adaptor Proteins, Signal Transducing/genetics , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Cell Shape/drug effects , Culture Media, Conditioned/metabolism , DNA-Binding Proteins/genetics , Fibroblast Growth Factor 2/pharmacology , Gene Expression Regulation , Histamine/pharmacology , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Muscle Proteins/genetics , Placenta Growth Factor/pharmacology , Platelet-Derived Growth Factor/pharmacology , Signal Transduction , TEA Domain Transcription Factors , Transcription Factors/genetics , Vascular Endothelial Growth Factors/pharmacology , YAP-Signaling Proteins
4.
Microvasc Res ; 128: 103934, 2020 03.
Article in English | MEDLINE | ID: mdl-31654655

ABSTRACT

Pathological angiogenesis is a hallmark of many diseases. Previously, we reported that orphan nuclear receptor TR3/Nur77 was a critical mediator of angiogenesis to regulate tumor growth, sepsis and skin wound healing. However, none of the TR3/Nur77 targeting molecule has been in clinical trial so far. Here, we designed and generated novel TR3 shRNAs and two minigenes that had therapeutic potential for cancer treatment. In addition to extend our previous findings that tumor growth was inhibited in Nur77 knockout mice, we found that metastasis of colorectal tumor was completely inhibited in Nur77-/- mice. Tumor masses were increased ~70% and decreased ~40% in our transgenic EC-Nur77-S mice and EC-Nur77-DN mice, in which the full-length cDNA and the dominant negative mutant of TR3/Nur77 were inducibly and specifically expressed in mouse endothelium, respectively. TR3 was highly expressed in the vasculature and tumor cells of human melanoma and colorectal cancer tissues, but not in normal tissues. The novel TR3 shRNAs and two minigenes almost completely inhibited the proliferation and migration of HUVECs and human melanoma A375sm cells. Angiogenesis induced by adenoviruses expressing VEGF and melanoma growth in mice were greatly and significantly inhibited by systemically administration of adenoviruses expressing TR3 shRNAs and two minigenes. Tumor angiogenesis and the expressions of genes associated with angiogenesis were greatly regulated in tumor tissues treated with TR3 shRNAs and minigenes. Taken together, these studies demonstrated that TR3/Nur77 was a specific therapeutic target for several human cancers by targeting both tumor cells and tumor microenvironment. These TR3/Nur77 biologics inhibit angiogenesis and tumor growth, and have translational potential.


Subject(s)
Neoplasms/therapy , Neovascularization, Pathologic , Neovascularization, Physiologic , Nuclear Receptor Subfamily 4, Group A, Member 1/metabolism , RNA, Small Interfering/genetics , RNAi Therapeutics , Animals , Cell Line, Tumor , Cell Movement , Cell Proliferation , Female , Gene Expression Regulation, Neoplastic , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Male , Mice, Inbred C57BL , Mice, Knockout , Mice, Nude , Neoplasm Metastasis , Neoplasms/genetics , Neoplasms/metabolism , Neoplasms/pathology , Nuclear Receptor Subfamily 4, Group A, Member 1/deficiency , Nuclear Receptor Subfamily 4, Group A, Member 1/genetics , RNA, Small Interfering/metabolism , Tumor Burden , Tumor Microenvironment , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/metabolism , Xenograft Model Antitumor Assays
5.
Am J Nephrol ; 51(10): 806-814, 2020.
Article in English | MEDLINE | ID: mdl-32966971

ABSTRACT

BACKGROUND: Cardiovascular outcomes in clinical trials with type 2 diabetes mellitus (T2DM) patients have shown that glucagon-like peptide-1 receptor agonist can have a beneficial effect on the kidney. This trial aimed to assess the effects of exenatide on renal outcomes in patients with T2DM and diabetic kidney disease (DKD). METHODS: We performed a randomized parallel study encompassing 4 general hospitals. T2DM patients with an estimated glomerular filtration rate (eGFR) ≥30 mL/min/1.73 m2 and macroalbuminuria, defined as 24-h urinary albumin excretion rate (UAER) >0.3 g/24 h were randomized 1:1 to receive exenatide twice daily plus insulin glargine (intervention group) or insulin lispro plus glargine (control group) for 24 weeks. The primary outcome was the UAER percentage change from the baseline after 24 weeks of intervention. The rates of hypoglycemia, adverse events (AEs), and change in eGFR during the follow-up were measured as safety outcomes. RESULTS: Between March 2016 and April 2019, 92 patients were randomized and took at least 1 dose of the study drug. The mean age of the participants was 56 years. At baseline, the median UAER was 1,512.0 mg/24 h and mean eGFR was 70.4 mL/min/1.73 m2. After 24 weeks of treatment, the UAER percentage change was significantly lower in the intervention group than in the control group (p = 0.0255). Moreover, the body weight declined by 1.3 kg in the intervention group (the difference between the 2 groups was 2.7 kg, p = 0.0001). Compared to the control group, a lower frequency of hypoglycemia and more gastrointestinal AEs were observed in the intervention group. CONCLUSION: Exenatide plus insulin glargine treatment for 24 weeks resulted in a reduction of albuminuria in T2DM patients with DKD.


Subject(s)
Albuminuria/drug therapy , Diabetes Mellitus, Type 2/drug therapy , Diabetic Nephropathies/drug therapy , Exenatide/administration & dosage , Hypoglycemic Agents/administration & dosage , Albuminuria/blood , Albuminuria/diagnosis , Albuminuria/etiology , Blood Glucose/analysis , Blood Glucose/drug effects , Diabetes Mellitus, Type 2/complications , Diabetic Nephropathies/blood , Diabetic Nephropathies/diagnosis , Diabetic Nephropathies/etiology , Disease Progression , Drug Therapy, Combination/adverse effects , Drug Therapy, Combination/methods , Exenatide/adverse effects , Female , Follow-Up Studies , Humans , Hypoglycemia/blood , Hypoglycemia/chemically induced , Hypoglycemia/diagnosis , Hypoglycemia/epidemiology , Hypoglycemic Agents/adverse effects , Insulin Glargine/administration & dosage , Insulin Glargine/adverse effects , Male , Middle Aged , Treatment Outcome
6.
Microvasc Res ; 124: 67-75, 2019 07.
Article in English | MEDLINE | ID: mdl-30930165

ABSTRACT

Pathological angiogenesis is a hallmark of many diseases. Previously, we reported that orphan nuclear receptor TR3/Nur77 was a critical mediator of angiogenesis to regulate tumor growth and skin wound healing via regulating the expression of the junctional proteins and integrins. However, the molecular mechanism, by which TR3/Nur77 regulates angiogenesis is not completely understood. Here, we were the first to find that TR3/Nur77, via its various amino acid fragments, regulated the expression of DLL4 and Jagged 1 in cultured endothelial cells. DLL4 and Jagged1 mediated TR3/Nur77-induced angiogenic responses and signaling molecules, but not the expression of integrins. Instead, integrins regulated the expressions of DLL4 and Jagged1 induced by TR3/Nur77. Further, DLL4, Jagged1 and integrins α1, α2, ß3 and ß5 were regulated by TR3/Nur77 in animal sepsis models of lipopolysaccharide (LPS)-induced endotoxemia, and cecal ligation and puncture (CLP), in which, TR3/Nur77 expression was significantly and tranciently increased. Mouse survival rates were greatly increased in Nur77 knockout mice bearing both CLP and LPS models. The results elucidated a novel axis of VEGF/histamine ➔ TR3/Nur77 ➔ integrins ➔ DLL4/Jagged1 in angiogenesis, and demonstrated that TR3/Nur77 was an excellent target for sepsis. These studies supported our previous findings that TR3/Nur77 was an excellent therapeutic target, and further our understanding of the molecular mechanism, by which TR3/Nur77 regulated angiogenesis.


Subject(s)
Endotoxemia/metabolism , Human Umbilical Vein Endothelial Cells/metabolism , Intercellular Signaling Peptides and Proteins/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Jagged-1 Protein/metabolism , Membrane Proteins/metabolism , Neovascularization, Physiologic , Nuclear Receptor Subfamily 4, Group A, Member 1/metabolism , Adaptor Proteins, Signal Transducing , Animals , Calcium-Binding Proteins , Cells, Cultured , Disease Models, Animal , Endotoxemia/genetics , Endotoxemia/pathology , Female , Humans , Integrins/metabolism , Male , Mice, Knockout , Neovascularization, Pathologic , Nuclear Receptor Subfamily 4, Group A, Member 1/deficiency , Nuclear Receptor Subfamily 4, Group A, Member 1/genetics , Signal Transduction
7.
Microvasc Res ; 122: 22-33, 2019 03.
Article in English | MEDLINE | ID: mdl-30391133

ABSTRACT

Pathological angiogenesis is a hallmark of many diseases. Previously, we reported that orphan nuclear receptor TR3/Nur77 (human homolog, Nur77, mouse homolog) is a critical mediator of angiogenesis to regulate tumor growth and skin wound healing via down-regulating the expression of the junctional proteins and integrin ß4. However, the molecular mechanism, by which TR3/Nur77 regulated angiogenesis, was still not completely understood. In this report by analyzing the integrin expression profile in endothelial cells, we found that the TR3/Nur77 expression highly increased the expression of integrins α1 and ß5, decreased the expression of integrins α2 and ß3, but had some or no effect on the expression of integrins αv, α3, α4, α5, α6, ß1 and ß7. In the angiogenic responses mediated by TR3/Nur77, integrin α1 regulated endothelial cell proliferation and adhesion, but not migration. Integrin ß5 shRNA inhibited cell migration, but increased proliferation and adhesion. Integrin α2 regulated all of the endothelial cell proliferation, migration and adhesion. However, integrin ß3 did not play any role in endothelial cell proliferation, migration and adhesion. TR3/Nur77 regulated the transcription of integrins α1, α2, ß3 and ß5, via various amino acid fragments within its transactivation domain and DNA binding domain. Furthermore, TR3/Nur77 regulated the integrin α1 promoter activity by directly interacting with a novel DNA element within the integrin α1 promoter. These studies furthered our understanding of the molecular mechanism by which TR3/Nur77 regulated angiogenesis, and supported our previous finding that TR3/Nur77 was an excellent therapeutic target for pathological angiogenesis. Therefore, targeting TR3/Nur77 inhibits several signaling pathways that are activated by various angiogenic factors.


Subject(s)
Endothelial Cells/metabolism , Integrins/metabolism , Microvessels/metabolism , Neovascularization, Physiologic , Nuclear Receptor Subfamily 4, Group A, Member 1/metabolism , Skin/blood supply , Angiogenesis Inducing Agents/pharmacology , Binding Sites , Cell Adhesion , Cell Movement , Cell Proliferation , Cells, Cultured , Endothelial Cells/drug effects , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Integrins/genetics , Microvessels/cytology , Microvessels/drug effects , Neovascularization, Physiologic/drug effects , Nuclear Receptor Subfamily 4, Group A, Member 1/genetics , Promoter Regions, Genetic , Signal Transduction , Transcription, Genetic , Vascular Endothelial Growth Factor A/pharmacology
8.
Microvasc Res ; 105: 103-8, 2016 May.
Article in English | MEDLINE | ID: mdl-26860974

ABSTRACT

VEGF, upon binding to its endothelial cell specific receptors VEGF-R1 and VEGF-R2, can induce endothelial cell migration, proliferation and angiogenesis. However, the molecular mechanism of these effects still remains unclear. In this study, we investigated whether VEGF promotes human umbilical vascular endothelial cell (HUVEC) migration and proliferation through activator protein-1 transcription factor (AP-1) family. We first showed that VEGF induces immediate-early genes AP-1 family gene expression differentially with the profound induction of JunB (both mRNA and protein) under various conditions (PBS, DMSO or control adenoviruses). The increase in AP-1 mRNA expression occurs primarily at the transcriptional level. Inhibition of AP-1 DNA binding activity by adenovirus expressing a potent dominant negative form of c-Fos (Afos) significantly attenuated VEGF-induced HUVEC migration and proliferation and cyclin D1 expression. Knockdown of JunB with adenovirus expressing JunB shRNA reduces VEGF-induced JunB expression and attenuated HUVEC migration. However the shJunB-expressing virus has no effect on VEGF-induced cyclin D1 protein expression and proliferation. These results suggest that VEGF-induced endothelial migration is mediated primarily by induction of JunB whereas the promotion of endothelial proliferation by VEGF is mediated by JunB-independent AP-1 family members.


Subject(s)
Cell Movement/drug effects , Cell Proliferation/drug effects , Human Umbilical Vein Endothelial Cells/drug effects , Neovascularization, Physiologic/drug effects , Proto-Oncogene Proteins c-fos/metabolism , Transcription Factor AP-1/metabolism , Transcription Factors/metabolism , Vascular Endothelial Growth Factor A/pharmacology , Cells, Cultured , Cyclin D1/metabolism , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Proto-Oncogene Proteins c-fos/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Signal Transduction/drug effects , Time Factors , Transcription Factor AP-1/genetics , Transcription Factors/genetics , Transcription, Genetic , Transfection , Up-Regulation
9.
Tumour Biol ; 37(3): 3307-20, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26440050

ABSTRACT

TR3 has been reported to be an excellent target for angiogenesis therapies. We reported three TR3 transcript variant messenger RNAs (mRNAs) are expressed in human umbilical vein endothelial cell (HUVEC) and are differentially regulated by vascular endothelial growth factor (VEGF). TR3 transcript variant 1 (TR3-TV1) and variant 2 (TR3-TV2) encoding the same TR3 isoform 1 protein (TR3-iso1) that was named TR3 has been extensively studied. However, the function of TR3 isoform 2 protein (TR3-iso2) encoded by TR3 transcript variant 3 (TR3-TV3) is still not known. Here, we clone and express the novel TR3-iso2 protein and find that expression of TR3-iso2, in contrast to TR3-iso1, inhibits endothelial cell proliferation induced by VEGF-A, histamine, and phorbol-12-myristate-13-acetate (PMA). The differential function of TR3-iso2 correlates with the down-regulation of cyclin D1. However, TR3-iso2 plays similar roles in endothelial cell migration and monolayer permeability as TR3-iso1. We further demonstrate that several intracellular signaling pathways are involved in histamine-induced TR3 transcript variants, including histamine receptor H1-mediated phospholipase C (PLC)/calcium /calcineurin/protein kinase C (PKC)/protein kinase D (PKD) pathway and ERK pathway, as well as histamine receptor H3-mediated PKC-ERK pathway. Further, expressions of TR3-TV1, TR3-TV2, and TR3-TV3 by VEGF and histamine are regulated by different promoters, but not by their mRNA stability.


Subject(s)
Cell Movement/genetics , Cell Proliferation/genetics , Human Umbilical Vein Endothelial Cells/metabolism , Nuclear Receptor Subfamily 4, Group A, Member 1/genetics , Cell Movement/drug effects , Cell Proliferation/drug effects , Cells, Cultured , Gene Expression/drug effects , Gene Expression Profiling/methods , Histamine/pharmacology , Human Umbilical Vein Endothelial Cells/drug effects , Humans , Immunoblotting , Nuclear Receptor Subfamily 4, Group A, Member 1/metabolism , Promoter Regions, Genetic/genetics , Protein Isoforms/genetics , Protein Isoforms/metabolism , RNA Interference , Reverse Transcriptase Polymerase Chain Reaction , Tetradecanoylphorbol Acetate/pharmacology , Vascular Endothelial Growth Factor A/pharmacology
10.
FASEB J ; 29(1): 131-40, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25326539

ABSTRACT

Tissue repair/wound healing, in which angiogenesis plays an important role, is a critical step in many diseases including chronic wound, myocardial infarction, stroke, cancer, and inflammation. Recently, we were the first to report that orphan nuclear receptor TR3/Nur77 is a critical mediator of angiogenesis and its associated microvessel permeability. Tumor growth and angiogenesis induced by VEGF-A, histamine, and serotonin are almost completely inhibited in Nur77 knockout mice. However, it is not known whether TR3/Nur77 plays any roles in wound healing. In these studies, skin wound-healing assay was performed in 3 types of genetically modified mice having various Nur77 activities. We found that ectopic induction of Nur77 in endothelial cells of mice is sufficient to improve skin wound healing. Although skin wound healing in Nur77 knockout mice is comparable to the wild-type control mice, the process is significantly delayed in the EC-Nur77-DN mice, in which a dominant negative Nur77 mutant is inducibly and specifically expressed in mouse endothelial cells. By a loss-of-function assay, we elucidate a novel feed-forward signaling pathway, integrin ß4 → PI3K → Akt → FAK, by which TR3 mediates HUVEC migration. Furthermore, TR3/Nur77 regulates the expression of integrin ß4 by targeting its promoter activity. In conclusion, expression of TR3/Nur77 improves wound healing by targeting integrin ß4. TR3/Nur77 is a potential candidate for proangiogenic therapy. The results further suggest that TR3/Nur77 is required for pathologic angiogenesis but not for developmental/physiologic angiogenesis and that Nur77 and its family members play a redundant role in normal skin wound healing.


Subject(s)
Integrin beta4/metabolism , Nuclear Receptor Subfamily 4, Group A, Member 1/metabolism , Wound Healing/physiology , Animals , Cell Movement , Cell Proliferation , Human Umbilical Vein Endothelial Cells , Humans , Integrin beta4/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Neovascularization, Pathologic , Neovascularization, Physiologic , Nuclear Receptor Subfamily 4, Group A, Member 1/deficiency , Nuclear Receptor Subfamily 4, Group A, Member 1/genetics , Promoter Regions, Genetic , RNA, Small Interfering/genetics , Signal Transduction , Skin/injuries , Skin/metabolism , Skin/pathology , Up-Regulation , Wound Healing/genetics
11.
Blood ; 121(11): 2154-64, 2013 Mar 14.
Article in English | MEDLINE | ID: mdl-23315169

ABSTRACT

Angiogenesis plays an important role in cancer and in many other human diseases. Vascular endothelial growth factor-A (VEGF-A), the best known angiogenic factor, was originally discovered as a potent vascular permeability factor (VPF), suggesting that other vascular permeabilizing agents, such as histamine and serotonin, might also have angiogenic activity. We recently demonstrated that, like VEGF-A, histamine and serotonin up-regulate the orphan nuclear receptor and transcription factor TR3 (mouse homolog Nur77) and that TR3/Nur77 is essential for their vascular permeabilizing activities. We now report that histamine and serotonin are also angiogenic factors that, at low micromolar concentrations, induce endothelial cell proliferation, migration and tube formation in vitro, and angiogenesis in vivo. All of these responses are mediated through specific histamine and serotonin receptors, are independent of VEGF-A, and are directly dependent on TR3/Nur77. Initially, the angiogenic response closely resembled that induced by VEGF-A, with generation of "mother" vessels. However, after ~10 days, mother vessels began to regress as histamine and serotonin, unlike VEGF-A, up-regulated the potent angiogenesis inhibitor thrombospondin-1, thereby triggering a negative feedback loop. Thus, histamine and serotonin induce an angiogenic response that fits the time scale of acute inflammation.


Subject(s)
Histamine/pharmacology , Neovascularization, Physiologic/drug effects , Nuclear Receptor Subfamily 4, Group A, Member 1/metabolism , Nuclear Receptor Subfamily 4, Group A, Member 1/physiology , Serotonin/pharmacology , Thrombospondin 1/physiology , Animals , Capillary Permeability/drug effects , Cells, Cultured , Female , Human Umbilical Vein Endothelial Cells/drug effects , Human Umbilical Vein Endothelial Cells/metabolism , Human Umbilical Vein Endothelial Cells/physiology , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Nude , Neovascularization, Physiologic/genetics , Nuclear Receptor Subfamily 4, Group A, Member 1/genetics , Protein Processing, Post-Translational/drug effects , Protein Processing, Post-Translational/genetics , Thrombospondin 1/metabolism , Up-Regulation/drug effects , Up-Regulation/genetics
12.
FASEB J ; 28(10): 4524-33, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25016027

ABSTRACT

Angiogenesis is a hallmark of many diseases, including cancer, ischemic heart disease, inflammation, and others. It is well known that vascular endothelial growth factor (VEGF) is the most important angiogenic factor. Recently, we demonstrated that orphan nuclear receptor TR3 (mouse Nur77 and rat NGFI-B) plays critical roles in tumor growth and angiogenesis induced by VEGF-A in vitro and in vivo. However, the signaling pathways that mediate the expression of TR3 induced by VEGF are still not completely understood. Here we reported that 3 TR3 transcript variants (TR3-TVs) are expressed at differential levels, and regulated differentially in endothelial cells. While the expression of TR3-TV1 is relatively low, the expression of TR3-TV2 is up-regulated markedly, and the expression of TR3-TV3 is up-regulated moderately in endothelial cells induced by VEGF-A. The kinetics of the induction of these TR3-TVs is different. We also found that several signaling pathways, including calcium-PLC-PKC-PKD1 pathway, NF-κB pathway, and MAP kinase (ERK, p38, and JNK) pathways are important for VEGF-A-induced TR3-TV2 and TR3-TV3 mRNA induction. More important, we found that VEGF-A or VEGF-E, but not VEGF-B, nor placenta growth factor (PlGF), induces the phosphorylation of insulin-like growth factor-1 receptor (IGF-1R) and the interaction of VEGF receptor 2/kinase insert domain receptor (VEGFR2/KDR) with IGF-1R, which mediates the expression of TR3-TV2, but not TR3-TV3. Taking together, we demonstrate that TR3-TVs are differentially regulated by VEGF-A and identify a novel signaling pathway by which VEGF-A and VEGF-E, but neither VEGF-B, nor PlGF, induce the interaction of VEGFR2/KDR with IGF-1R, resulting in IGF-1R transactivation to induce the high level expression of TR3-TV2. Our data not only elucidate the signaling pathways by which TR3-TVs are regulated, but extend the molecular mechanism, by which VEGF-A-induced angiogenesis. These studies should permit the development of screening assays for compounds that inhibit VEGF signaling.


Subject(s)
MAP Kinase Signaling System , Nuclear Receptor Subfamily 4, Group A, Member 1/metabolism , Vascular Endothelial Growth Factor A/pharmacology , Calcium/metabolism , Human Umbilical Vein Endothelial Cells/drug effects , Human Umbilical Vein Endothelial Cells/metabolism , Humans , NF-kappa B/metabolism , Nuclear Receptor Subfamily 4, Group A, Member 1/genetics , Placenta Growth Factor , Pregnancy Proteins/pharmacology , Protein Isoforms/genetics , Protein Isoforms/metabolism , Protein Kinase C/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptor, IGF Type 1/metabolism , Vascular Endothelial Growth Factor Receptor-2/metabolism
13.
Proc Natl Acad Sci U S A ; 108(29): 12066-71, 2011 Jul 19.
Article in English | MEDLINE | ID: mdl-21730126

ABSTRACT

Low-level basal vascular permeability (BVP) provides nutrients to normal tissues, and increased vascular permeability is characteristic of inflammation and cancer. We recently reported that VEGF-A, a potent vascular permeabilizing and angiogenic factor, exerts much of its angiogenic activity by up-regulating expression of TR3/Nur77, an orphan nuclear transcription factor, in vascular endothelial cells (EC). To determine whether TR3/Nur77 had a more general role in regulating vascular permeability, we found that histamine, serotonin, and platelet-activating factor, small molecule vascular permeabilizing agents, also increased TR3/Nur77 expression acutely in EC. BVP and the acute vascular hyperpermeability (AVH) induced by these vascular permeabilizing factors were greatly decreased in Nur77(-/-) mice, and both BVP and AVH correlated with Nur77 expression levels in several different mouse strains. BVP and AVH were enhanced in transgenic mice in which Nur77 was selectively overexpressed in vascular EC, whereas both were suppressed in mice overexpressing dominant-negative Nur77. Chronic vascular hyperpermeability (CVH) was induced long before the onset of angiogenesis in a modified, in vivo Matrigel assay that included PT67 cells packaging retroviruses expressing Nur77-sense, whereas inclusion of cells packaging viruses expressing Nur77-antisense prevented VEGF-A-induced CVH. TR3/Nur77 modulated vascular permeability by increasing endothelial nitric-oxide synthase expression and by downregulating several EC junction proteins that maintain vascular homeostasis. Both functions required TR3/Nur77 transcriptional activity. Taking these data together, TR3/Nur77 is up-regulated by several vascular permeabilizing agents and has critical roles in mediating BVP, AVH, and CVH.


Subject(s)
Capillary Permeability/physiology , Gene Expression Regulation/physiology , Intercellular Junctions/physiology , Microvessels/physiology , Nitric Oxide Synthase Type III/metabolism , Nuclear Receptor Subfamily 4, Group A, Member 1/metabolism , Analysis of Variance , Animals , Blotting, Western , Cell Line , Collagen , DNA Primers/genetics , Drug Combinations , Immunohistochemistry , Laminin , Mice , Mice, Transgenic , Nuclear Receptor Subfamily 4, Group A, Member 1/genetics , Polymerase Chain Reaction , Proteoglycans
14.
Phytomedicine ; 133: 155874, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39079314

ABSTRACT

BACKGROUND: The presence of distant metastasis at the time of initial diagnosis is a prevalent issue in non-small cell lung cancer (NSCLC), affecting around 30-40 % of the patients. Acidic tumor microenvironment (TME) provides favorable conditions that increase the invasiveness and aggressiveness of NSCLC. The activity of the glycolytic enzyme lactate dehydrogenase (LDHA) increases intracellular lactate accumulation, which creates an acidic TME. However, it is not yet known whether LDHA is involved in enhancing the metastatic potential of NSCLC and if LDHA is a druggable therapeutic target for NSCLC. PURPOSE: We aimed to investigate the molecular mechanisms underlying the enhanced NSCLC metastasis in acidic TME, and to explore whether sulforaphane (SFN), an active compound in Raphani Semen, can serve as a LDHA inhibitor to inhibit NSCLC metastasis in the acidic TME. METHODS: To mimic the acidic TME, NSCLC cells were cultured in acidic medium (pH 6.6), normal medium (pH 7.4) served as control. Western blotting, bioinformatic analysis, luciferase assay and rescue experiments were used to explore the mechanism and investigate the anti-metastatic effect of SFN both in vitro and in vivo. RESULTS: Acidic environment increases the expression of LDHA which in turn increases the production of lactic acid that contributes to the acidity of TME. Interestingly, elevated LDHA expression results from increased c-Myc expression, which transactivates LDHA. c-Myc expression is directly regulated by miR-7-5p. In vitro study shows that overexpression of miR-7-5p reverses the acidic pH-enhanced c-Myc and LDHA expressions and also abolishes the enhanced NSCLC cell migration. More importantly, SFN significantly inhibits NSCLC growth and metastasis by reducing lactate production via the miR-7-5p/c-Myc/LDHA axis. Besides, it also regulates the expressions of monocarboxylate transporter 1 (MCT1) and MCT4 that transport lactate across cell membrane. CONCLUSIONS: The miR-7-5p/c-Myc/LDHA axis is involved in the enhanced NSCLC metastasis in the acidic TME. SFN, a novel LDHA inhibitor, reduces lactate production by targeting the miR-7-5p/c-Myc/LDHA axis, and hence inhibits NSCLC metastasis. Our findings not only delineate a novel mechanism, but also support the clinical translation of SFN as a novel therapeutic agent for treating metastatic NSCLC.


Subject(s)
Carcinoma, Non-Small-Cell Lung , Isothiocyanates , L-Lactate Dehydrogenase , Lung Neoplasms , MicroRNAs , Proto-Oncogene Proteins c-myc , Sulfoxides , Tumor Microenvironment , Isothiocyanates/pharmacology , Carcinoma, Non-Small-Cell Lung/drug therapy , Tumor Microenvironment/drug effects , Sulfoxides/pharmacology , MicroRNAs/metabolism , Humans , Lung Neoplasms/drug therapy , Animals , Proto-Oncogene Proteins c-myc/metabolism , Cell Line, Tumor , L-Lactate Dehydrogenase/metabolism , Mice, Nude , Mice , Hydrogen-Ion Concentration , Mice, Inbred BALB C , Neoplasm Metastasis , Cell Movement/drug effects
15.
ACS Appl Mater Interfaces ; 16(39): 52414-52422, 2024 Oct 02.
Article in English | MEDLINE | ID: mdl-39302810

ABSTRACT

The use of a bipolar membrane (BPM) in a hybrid acid/alkali electrolyzer is widely considered as a promising energy technology for efficient hydrogen production. The stability of a BPM is often believed to be largely limited by the anion exchange layer (AEL) due to the hydrophilic attack of AEL polymers by hydroxide groups in alkaline. In this study, we employ X-ray computed tomography (CT) to investigate the degradation behaviors of BPM and found that the cation exchange layer (CEL) experiences more pronounced degradation compared with the AEL during water splitting operations. Despite its chemical stability in both acidic and alkaline environments, the CEL is more prone to electrochemical corrosion under the influence of applied voltages. This susceptibility leads to the formation of micropores and a consequent increase in the porosity. The results of this work provide a new perspective on and highlight the complexity of the degradation behaviors of BPMs in hybrid acid/alkali electrolyzers.

16.
J Colloid Interface Sci ; 674: 108-117, 2024 Nov 15.
Article in English | MEDLINE | ID: mdl-38917711

ABSTRACT

Large-scale hydrogen production by electrocatalytic water splitting still remains as a critical challenge due to the severe catalyst degradation during the oxygen evolution reaction (OER) in acidic media. In this study, we investigate the structural impacts on catalyst degradation behaviors using three iridium-based oxides, namely SrIrO3, Sr2IrO4, and Sr4IrO6 as model catalysts. These Ir oxides possess different connection configurations of [IrO6] octahedra units in their structure. Stable OER performance is observed on SrIrO3 and attributed to the edge-linked [IrO6] structure and rapid formation of a continuous IrOx layer on its surface, which functions not only as the "real" catalyst but also a shield preventing continuous cation leaching (with <1.0 at.% of Ir leaching). In comparison, both Sr2IrO4 and Sr4IrO6 catalysts demonstrate quick current fading with structure transformation to rutile IrO2 and formation of inconducive SrSO4 precipitates on surface, blocking the reactive sites. Nevertheless, over 60 at.% of Ir leaching is detected from the Sr4IrO6 catalyst due to its isolated [IrO6] structure configuration. Results of this work highlight the structural impacts on the catalyst stability in acidic OER conditions.

17.
Mater Horiz ; 11(19): 4737-4746, 2024 Sep 30.
Article in English | MEDLINE | ID: mdl-39011761

ABSTRACT

Recently, polar side chains have emerged as a functional tool to enhance conjugated polymer doping properties by improving the polymer miscibility with polar chemical dopants and facilitate solvated ion uptake. In this work, we design and investigate a novel family of side chains containing a single ether function, enabling the modulation of the oxygen atom position along the side chain. A meticulous investigation of this new polymer series by differential scanning calorimetry, fast scanning chip calorimetry and X-ray scattering shows that polymers bearing single-ether side chains can show high degree of crystallinity under proper conditions. Importantly, due to a gauche effect allowing the side chain to bend at the oxygen atom, the degree of crystallinity of polymers can be controlled by the position of the oxygen atom along the side chain. The further the oxygen atom is from the conjugated backbone, the more crystalline the polymer becomes. In addition, for all new polymers, high thermomechanical properties are demonstrated, leading to remarkable electrical conductivities and thermoelectric power factors in rub-aligned and sequentially doped thin films. This work confirms the potential of single-ether side chains to be used as polar solubilizing side chains for the design of a next generation of p- and n-type semiconducting polymers with increased affinity to polar dopants while maintaining high molecular order.

18.
Chin J Integr Med ; 30(3): 195-202, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38374490

ABSTRACT

OBJECTIVE: To evaluate the effect and safety of foot baths with Tangbi Waixi Decoction (TW) in treating patients with diabetic peripheral neuropathy (DPN). METHODS: It is a multicenter double-blinded randomized controlled trial. Participants with DPN were recruited between November 18, 2016 and May 30, 2018 from 8 hospitals in China. All patients received basic treatments for glycemic management. Patients received foot baths with TW herbal granules either 66.9 g (intervention group) or 6.69 g (control group) for 30 min once a day for 2 weeks and followed by a 2-week rest, as a therapeutic course. If the Toronto Clinical Scoring System total score (TCSS-TS) ⩾6 points, the patients received a total of 3 therapeutic courses (for 12 weeks) and were followed up for 12 weeks. The primary outcome was change in TCSS-TS score at 12 and 24 weeks. Secondary outcomes included changes in bilateral motor nerve conduction velocity (MNCV) and sensory nerve conduction velocity (SNCV) of the median and common peroneal nerve. Safety was also assessed. RESULTS: Totally 632 patients were enrolled, and 317 and 315 were randomized to the intervention and control groups, respectively. After the 12-week intervention, patients in both groups showed significant declines in TCSSTS scores, and significant increases in MNCV and SNCV of the median and common peroneal nerves compared with pre-treatment (P<0.05). The reduction of TCSS-TS score at 12 weeks and the increase of SNCV of median nerve at 24 weeks in the control group were greater than those in the intervention group (P<0.05). The number of adverse events did not differ significantly between groups (P>0.05), and no serious adverse event was related with treatment. CONCLUSION: Treatment of TW foot baths was safe and significantly benefitted patients with DPN. A low dose of TW appeared to be more effective than a high dose. (Registry No. ChiCTR-IOR-16009331).


Subject(s)
Diabetes Mellitus , Diabetic Neuropathies , Plants, Medicinal , Humans , Diabetic Neuropathies/drug therapy , Baths , Double-Blind Method , Plant Extracts/therapeutic use
19.
Arterioscler Thromb Vasc Biol ; 32(7): 1707-16, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22628435

ABSTRACT

OBJECTIVE: Vascular endothelial growth factor (VEGF) promotes leukocyte adhesion to endothelial cells (ECs). Angiopoietin-1 (Ang-1) inhibits this response. Nuclear receptor-77 (Nur77) is a proangiogenic nuclear receptor. In the present study, we assessed the influence of Ang-1 and VEGF on Nur77 expression in ECs, and evaluated its role in Ang-1/VEGF-mediated leukocyte adhesion. METHODS AND RESULTS: Expression of Nur77 was evaluated with real-time polymerase chain reaction and immunoblotting. Adhesion of leukocytes to ECs was monitored with inverted microscopy. Nur77 expression or activity was inhibited using adenoviruses expressing dominant-negative form of Nur77, retroviruses expressing Nur77 in the antisense direction, and small interfering RNA oligos. Both Ang-1 and VEGF induce Nur77 expression, by >5- and 30-fold, respectively. When combined, Ang-1 potentiates VEGF-induced Nur77 expression. Ang-1 induces Nur77 through the phosphoinositide 3-kinase and extracellular signal-regulated protein kinase 1/2 pathways. VEGF induces Nur77 expression through the protein kinase D/histone deacetylase 7/myocyte enhancer factor 2 and extracellular signal-regulated protein kinase 1/2 pathways. VEGF induces nuclear factor-kappaB transcription factor, vascular cell adhesion molecule-1, and E-selectin expressions, and promotes leukocyte adhesion to ECs. Ang-1 inhibits these responses. This inhibitory effect of Ang-1 disappears when Nur77 expression is disrupted, restoring the inductive effects of VEGF on adhesion molecule expression, and increased leukocyte adhesion to ECs. CONCLUSIONS: Nur77 promotes anti-inflammatory effects of Ang-1, and functions as a negative feedback inhibitor of VEGF-induced EC activation.


Subject(s)
Angiopoietin-1/pharmacology , Endothelial Cells/drug effects , Leukocytes/drug effects , Nuclear Receptor Subfamily 4, Group A, Member 1/physiology , Vascular Endothelial Growth Factor A/pharmacology , Cell Adhesion/drug effects , Cells, Cultured , Endothelial Cells/physiology , Histone Deacetylases/metabolism , Humans , I-kappa B Kinase/genetics , Leukocytes/physiology , NF-kappa B/metabolism , Nuclear Receptor Subfamily 4, Group A, Member 1/genetics , Phosphatidylinositol 3-Kinases/physiology , Phosphorylation , Proto-Oncogene Proteins c-akt/physiology , TRPP Cation Channels/metabolism , U937 Cells , Vascular Cell Adhesion Molecule-1/genetics
20.
ACS Appl Mater Interfaces ; 15(12): 15269-15278, 2023 Mar 29.
Article in English | MEDLINE | ID: mdl-36930828

ABSTRACT

Even the most stable Ir-based oxides inevitably encounter a severe degradation problem during the oxygen evolution reaction (OER) in acid, resulting in quick formation of amorphous IrOx layers on the catalyst surface. Unfortunately, there is still a lack of fundamental understanding of such hydrous IrOx layers, including the atomic arrangement, key active structure, compositions, chemical stability, and so on. In this work, we demonstrate an electrochemical strategy to prepare two types of protonated iridium oxides with well-defined crystalline structures: one possesses a 2D layered structure (denoted as α-HxIrO3) and the other consists of 3D interconnected polymorphs (denoted as ß-HxIrO3). Both protonated iridium oxides demonstrate superior electrochemical stabilities with 6 times suppressed Ir dissolution comparing to the initial Li2IrO3 and rutile IrO2. It is hypothesized that the enriched protons and fast diffusions in these two protonated HxIrO3 crystal oxides may promote surface structural stability by suppressing the formation of high-valence Ir species at the solid-liquid interfaces during OER. Overall, the results of this work shed light on the role of proton dynamics toward the OER processes on the catalyst surface in acid media.

SELECTION OF CITATIONS
SEARCH DETAIL