Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 48
Filter
1.
Acta Histochem ; 125(8): 152100, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37837833

ABSTRACT

OBJECTIVE: Idiopathic pulmonary fibrosis (IPF) is considered as a chronic interstitial lung disease with underlying mechanism of IPF remaining unclear, while there are no definitive treatment options. In recent years, scientists have gradually paid attention to the influence of angiogenesis on IPF. Because IPF is a progressive with microvascular remodeling disorder, scientists have postulated that angiogenesis may also be one of the initiating and contributing factors of the disease. Bupleurum is a common natural Chinese herbal medicine with antibacterial, anti-inflammatory, anti-tumor and other pharmacological effects. As the most important active monomer of Bupleurum, Saikosaponin-d (SSd) is a new discovery with anti-pulmonary fibrosis (PF) activity. This study attempts to investigate the role of SSd in the interference of PF through regulation of angiogenesis in IPF through Angiopoietin (Angpt) /Tie receptor 2 (Tie2) pathway. METHODS: Randomly, we allocated C57BL/6 mice into four groups (n = 20 in each group). Afterwards, establishment of IPF model was accomplished via intratracheal administration of bleomycin (BLM, 5 mg/kg), while corresponding drug intervention was given accordingly. On 3rd, 7th, 14th and 28th days after modeling, we performed histopathological examination through staining. Meanwhile, immunohistochemistry (IHC) of PF and the expression of related factors were observed, while Ang/Tie2 pathway was assessed by ELISA with the effect of SSd on angiogenesis related proteins in IPF being explored with IHC and Western Blot technique. RESULTS: Our results showed that SSd could reduce inflammation and PF levels in lung tissue of experimental mice, while levels of angiogenesis-related factors, namely Tie-2, Ang-1 and ANGPT2 (Ang-2), fibrosis- associated factors like Alpha-smooth muscle actin (α-SMA), collagen-I and hydroxyproline in SSd and dexamethasone (DXM) mice were significantly reduced at each time point compared to BLM (p < 0.01). Additionally, we discovered substantial decreased expressions of Ang-1, Ang-2, Tie-2, α-SMA and collagen-I at protein level in SSd and DXM mice at each time point compared to BLM (p < 0.05). Besides, insignificant differences were observed between SSd and DXM groups (p > 0.05). CONCLUSION: This study has demonstrated that SSd could down-regulate the expression of ANG-1, Ang-2 and Tie2 in the Ang/Tie2 pathway, and may reduce lung inflammation and PF in BLM-induced mice via inhibition of angiogenesis.


Subject(s)
Angiopoietins , Idiopathic Pulmonary Fibrosis , Mice , Animals , Angiopoietins/metabolism , Angiopoietins/pharmacology , Mice, Inbred C57BL , Lung/metabolism , Idiopathic Pulmonary Fibrosis/drug therapy , Idiopathic Pulmonary Fibrosis/metabolism , Idiopathic Pulmonary Fibrosis/pathology , Collagen Type I/metabolism , Bleomycin/pharmacology , Bleomycin/metabolism
2.
J Exp Med ; 220(1)2023 01 02.
Article in English | MEDLINE | ID: mdl-36269299

ABSTRACT

Primary tumors and distant site metastases form a bidirectionally communicating system. Yet, the molecular mechanisms of this crosstalk are poorly understood. Here, we identified the proteolytically cleaved fragments of angiopoietin-like 4 (ANGPTL4) as contextually active protumorigenic and antitumorigenic contributors in this communication ecosystem. Preclinical studies in multiple tumor models revealed that the C-terminal fragment (cANGPTL4) promoted tumor growth and metastasis. In contrast, the N-terminal fragment of ANGPTL4 (nANGPTL4) inhibited metastasis and enhanced overall survival in a postsurgical metastasis model by inhibiting WNT signaling and reducing vascularity at the metastatic site. Tracing ANGPTL4 and its fragments in tumor patients detected full-length ANGPTL4 primarily in tumor tissues, whereas nANGPTL4 predominated in systemic circulation and correlated inversely with disease progression. The study highlights the spatial context of the proteolytic cleavage-dependent pro- and antitumorigenic functions of ANGPTL4 and identifies and validates nANGPTL4 as a novel biomarker of tumor progression and antimetastatic therapeutic agent.


Subject(s)
Angiopoietin-Like Protein 4 , Neoplasms , Humans , Angiopoietin-Like Protein 4/pharmacology , Angiopoietin-Like Protein 4/therapeutic use , Angiopoietins/pharmacology , Angiopoietins/therapeutic use , Biomarkers, Tumor , Neoplasms/diagnosis , Neoplasms/drug therapy , Neoplasms/metabolism , Peptide Fragments/pharmacology , Peptide Fragments/therapeutic use
3.
Nat Med ; 28(12): 2633-2645, 2022 12.
Article in English | MEDLINE | ID: mdl-36456835

ABSTRACT

Osteoarthritis (OA) is a common, debilitating, chronic disease with no disease-modifying drug approved to date. We discovered LNA043-a derivative of angiopoietin-like 3 (ANGPTL3)-as a potent chondrogenesis inducer using a phenotypic screen with human mesenchymal stem cells. We show that LNA043 promotes chondrogenesis and cartilage matrix synthesis in vitro and regenerates hyaline articular cartilage in preclinical OA and cartilage injury models in vivo. LNA043 exerts at least part of these effects through binding to the fibronectin receptor, integrin α5ß1 on mesenchymal stem cells and chondrocytes. In a first-in-human (phase 1), randomized, double-blinded, placebo-controlled, single ascending dose, single-center trial ( NCT02491281 ; sponsored by Novartis Pharmaceuticals), 28 patients with knee OA were injected intra-articularly with LNA043 or placebo (3:1 ratio) either 2 h, 7 d or 21 d before total knee replacement. LNA043 met its primary safety endpoint and showed short serum pharmacokinetics, cartilage penetration and a lack of immunogenicity (secondary endpoints). Post-hoc transcriptomics profiling of cartilage revealed that a single LNA043 injection reverses the OA transcriptome signature over at least 21 d, inducing the expression of hyaline cartilage matrix components and anabolic signaling pathways, while suppressing mediators of OA progression. LNA043 is a novel disease-modifying OA drug candidate that is currently in a phase 2b trial ( NCT04864392 ) in patients with knee OA.


Subject(s)
Cartilage, Articular , Osteoarthritis, Knee , Humans , Osteoarthritis, Knee/drug therapy , Chondrocytes , Signal Transduction , Angiopoietins/metabolism , Angiopoietins/pharmacology , Angiopoietins/therapeutic use , Angiopoietin-Like Protein 3
4.
Allergol Immunopathol (Madr) ; 50(5): 47-56, 2022.
Article in English | MEDLINE | ID: mdl-36086963

ABSTRACT

It has been found that angiopoietin-like 4 (ANGPTL4) expression is increased in the serum of patients with chronic obstructive pulmonary disease (COPD). Herein, cigarette smoke extract (CSE) was used to stimulate oxidative stress in bronchial epithelial cells BEAS-2B, and the role and potential mechanism of ANGPTL4 in smoking-induced lung dysfunction were explored. The roles of different concentrations of CSE (0, 1, 2.5, 5, or 10%) in cell viability and ANGPTL4 levels were evaluated. Following ANGPTL4 being knocked down, the effects of ANGPTL4 knockdown on oxidative stress and apoptosis were determined. Moreover, the level of NADPH oxidase 2 (NOX2) was upregulated to assess the mediated role of NOX in the regulation of ANGPTL4, along with JNK/p38 MAPK signaling. CSE treatment elevated the level of ANGPTL4, and ANGPTL4 knockdown reduced CSE-induced oxidative stress, apoptosis, and NOX level in BEAS-2B cells. The greatest degree of alteration was found in NOX2, and additional NOX2 overexpression broke the inhibitory influences of ANGPTL4 knockdown on oxidative stress and apoptosis. Otherwise, ANGPTL4 knockdown hindered the activation of JNK/p38 MAPK signaling, whereas NOX2 overexpression activated this signaling pathway. Together, ANGPTL4 knockdown attenuated CSE-induced oxidative stress, apoptosis, and activation of JNK/MAPK signaling by inhibiting NOX.


Subject(s)
Cigarette Smoking , NADPH Oxidases , Angiopoietins/metabolism , Angiopoietins/pharmacology , Apoptosis , Cell Line , Cigarette Smoking/adverse effects , Epithelial Cells/metabolism , Humans , Lung , NADPH Oxidases/genetics , Oxidative Stress , p38 Mitogen-Activated Protein Kinases/metabolism
5.
Int J Biol Macromol ; 221: 1428-1438, 2022 Nov 30.
Article in English | MEDLINE | ID: mdl-36122781

ABSTRACT

Angiogenesis forms new vessels from existing ones. Abnormal angiogenesis, which is what gives tumor microenvironments their distinctive features, is characterised by convoluted, permeable blood vessels with a variety of shapes and high perfusion efficiency. Tumor angiogenesis controls cancer growth by allowing invasion and metastasis and is highly controlled by signalling networks. Therapeutic techniques targeting VEGF, PDGF, FGF Notch, Angiopoietin, and HGF signalling restrict the tumor's vascular supply. Numerous pathways regulate angiogenesis, and when one of those processes is blocked, the other pathways may step in to help. VEGF signalling inhibition alone has limits as an antiangiogenic therapy, and additional angiogenic pathways such as FGF, PDGF, Notch, angiopoietin, and HGF are important. For the treatment of advanced solid tumors, there are also new, emerging medicines that target multiple angiogenic pathways. Recent therapies block numerous signalling channels concurrently. This study focuses on 'alternative' methods to standard antiangiogenic medicines, such as cyclooxygenase-2 blocking, oligonucleotide binding complementary sites to noncoding RNAs to regulate mRNA target, matrix metalloproteinase inhibition and CRISPR/Cas9 based gene edition and dissecting alternative angiogenesis mechanism in tumor microenvironment.


Subject(s)
Neoplasms , Vascular Endothelial Growth Factor A , Humans , Vascular Endothelial Growth Factor A/metabolism , Angiopoietins/pharmacology , Angiopoietins/therapeutic use , Neovascularization, Pathologic/drug therapy , Signal Transduction , Angiogenesis Inhibitors/pharmacology , Neoplasms/metabolism , Tumor Microenvironment , Hepatocyte Growth Factor/therapeutic use
6.
Mol Biol Rep ; 48(1): 941-950, 2021 Jan.
Article in English | MEDLINE | ID: mdl-33393005

ABSTRACT

Tissue engineering is a rapidly developing field with many potential clinical applications in tissue and organ regeneration. The development of a mature and stable vasculature within these engineered tissues (ET) remains a significant obstacle. Currently, several growth factors (GFs) have been identified to play key roles within in vivo angiogenesis, including vascular endothelial growth factor (VEGF), platelet derived growth factor (PDGF), FGF and angiopoietins. In this article we attempt to build on in vivo principles to review the single, dual and multiple GF release systems and their effects on promoting angiogenesis. We conclude that multiple GF release systems offer superior results compared to single and dual systems with more stable, mature and larger vessels produced. However, with more complex release systems this raises other problems such as increased cost and significant GF-GF interactions. Upstream regulators and pericyte-coated scaffolds could provide viable alternative to circumnavigate these issues.


Subject(s)
Blood Vessels/drug effects , Fibroblast Growth Factors/pharmacology , Neovascularization, Physiologic/genetics , Tissue Engineering/methods , Vascular Endothelial Growth Factor A/pharmacology , Angiopoietins/genetics , Angiopoietins/metabolism , Angiopoietins/pharmacology , Animals , Blood Vessels/cytology , Blood Vessels/growth & development , Blood Vessels/metabolism , Endothelial Cells/cytology , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Fibroblast Growth Factors/genetics , Fibroblast Growth Factors/metabolism , Gene Expression Regulation , Humans , Matrix Metalloproteinases/genetics , Matrix Metalloproteinases/metabolism , Matrix Metalloproteinases/pharmacology , Pericytes/cytology , Pericytes/drug effects , Pericytes/metabolism , Platelet-Derived Growth Factor/genetics , Platelet-Derived Growth Factor/metabolism , Platelet-Derived Growth Factor/pharmacology , Regeneration/drug effects , Regeneration/genetics , Tissue Scaffolds , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/metabolism
7.
Neuroscience ; 411: 177-184, 2019 07 15.
Article in English | MEDLINE | ID: mdl-31152935

ABSTRACT

Angiopoietin (Ang) is an angiogenic factor, but its neuroprotective and neurotrophic effects have recently come to light. Ang exerts neuroprotective effects by inhibiting neuronal apoptosis, protecting the blood-brain/blood-spinal cord barrier, reducing inflammation and promoting neovascularization. In addition, Ang can also promote neural development and neurite outgrowth via activation of the PI3K/Akt signaling pathway and binding to the Tie2 receptor and/or integrin receptor. In addition, Ang and vascular endothelial growth factor (VEGF) are known to interact in blood vessels in the nervous system and the combination of Ang and VEGF can mitigate the negative effects of VEGF, such as inflammation and local edema. These data indicated that Ang is a novel neuroprotective/neurotrophic factor, which may become a new tool for the treatment of nerve injury.


Subject(s)
Angiopoietins/therapeutic use , Nerve Regeneration/drug effects , Neuroprotective Agents/therapeutic use , Peripheral Nerve Injuries/drug therapy , Angiopoietins/pharmacology , Animals , Humans , Neovascularization, Physiologic/drug effects , Neuroprotective Agents/pharmacology
8.
PLoS One ; 11(7): e0159276, 2016.
Article in English | MEDLINE | ID: mdl-27410263

ABSTRACT

The ß-cell mitogenic effects of ANGPTL8 have been subjected to substantial debate. The original findings suggested that ANGPTL8 overexpression in mice induced a 17-fold increase in ß-cell proliferation. Subsequent studies in mice contested this claim, but a more recent report in rats supported the original observations. These conflicting results might be explained by variable ANGPTL8 expression and differing methods of ß-cell quantification. To resolve the controversy, three independent labs collaborated on a blinded study to test the effects of ANGPTL8 upon ß-cell proliferation. Recombinant human betatrophin (hBT) fused to maltose binding protein (MBP) was delivered to mice by intravenous injection. The results demonstrate that ANGPTL8 does not stimulate significant ß-cell proliferation. Each lab employed different methods for ß-cell identification, resulting in variable quantification of ß-cell proliferation and suggests a need for standardizing practices for ß-cell quantification. We also observed a new action of ANGPTL8 in stimulating CD45+ hematopoietic-derived cell proliferation which may explain, in part, published discrepancies. Overall, the hypothesis that ANGPTL8 induces dramatic and specific ß-cell proliferation can no longer be supported. However, while ANGPTL8 does not stimulate robust ß-cell proliferation, the original experimental model using drug-induced (S961) insulin resistance was validated in subsequent studies, and thus still represents a robust system for studying signals that are either necessary or sufficient for ß-cell expansion. As an added note, we would like to commend collaborative group efforts, with repetition of results and procedures in multiple laboratories, as an effective method to resolve discrepancies in the literature.


Subject(s)
Angiopoietins/pharmacology , B-Lymphocytes/metabolism , Cell Proliferation/drug effects , Maltose-Binding Proteins/pharmacology , Mitogens/pharmacology , Peptide Hormones/pharmacology , Recombinant Proteins/pharmacology , Angiopoietin-Like Protein 8 , Angiopoietin-like Proteins , Angiopoietins/metabolism , Animals , Cells, Cultured , Male , Mice
9.
PLoS One ; 11(2): e0148564, 2016.
Article in English | MEDLINE | ID: mdl-26844767

ABSTRACT

Severe dengue is caused by host responses to viral infection, but the pathogenesis remains unknown. This is, in part, due to the lack of suitable animal models. Here, we report a non-mouse-adapted low-passage DENV-3 clinical isolate, DV3P12/08, derived from recently infected patients. DV3P12/08 caused a lethal systemic infection in type I and II IFN receptor KO mice (IFN-α/ß/γR KO mice), which have the C57/BL6 background. Infection with DV3P12/08 induced a cytokine storm, resulting in severe vascular leakage (mainly in the liver, kidney and intestine) and organ damage, leading to extensive hemorrhage and rapid death. DV3P12/08 infection triggered the release of large amounts of TNF-α, IL-6, and MCP-1. Treatment with a neutralizing anti-TNF-α antibody (Ab) extended survival and reduced liver damage without affecting virus production. Anti-IL-6 neutralizing Ab partly prolonged mouse survival. The anti-TNF-α Ab suppressed IL-6, MCP-1, and IFN-γ levels, suggesting that the severe response to infection was triggered by TNF-α. High levels of TNF-α mRNA were expressed in the liver and kidneys, but not in the small intestine, of infected mice. Conversely, high levels of IL-6 mRNA were expressed in the intestine. Importantly, treatment with Angiopoietin-1, which is known to stabilize blood vessels, prolonged the survival of DV3P12/08-infected mice. Taken together, the results suggest that an increased level of TNF-α together with concomitant upregulation of Tie2/Angiopoietin signaling have critical roles in severe dengue infection.


Subject(s)
Angiopoietins/metabolism , Capillary Permeability , Dengue Virus , Dengue/genetics , Dengue/metabolism , Receptor, TIE-2/metabolism , Receptors, Interferon/deficiency , Signal Transduction , Tumor Necrosis Factor-alpha/metabolism , Acute Disease , Angiopoietins/pharmacology , Animals , Antibodies, Monoclonal/pharmacology , Antibodies, Neutralizing/pharmacology , Biomarkers , Cytokines/blood , Cytokines/genetics , Cytokines/metabolism , Dengue/mortality , Dengue/virology , Disease Models, Animal , Gene Expression , Inflammation Mediators/blood , Inflammation Mediators/metabolism , Liver/metabolism , Liver/pathology , Liver Function Tests , Mice , Mice, Knockout , Thrombocytopenia/etiology , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Tumor Necrosis Factor-alpha/genetics , Viral Load
10.
Biochim Biophys Acta ; 1861(2): 130-137, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26569053

ABSTRACT

AIM: Atypical angiopoietin-like 8 (ANGPTL8), also known as betatrophin, is known to regulate lipid metabolism. However, its mechanism of action remains elusive. METHODS: HepG2, 3T3-L1, and NIT-1 cells were cultured in amino acid-complete MEM or histidine-free MEM to detect ANGPTL8 expression. The three cell types were treated with or without recombinant ANGPTL8 to investigate its role in lipid metabolism. Hydrodynamic tail vein gene delivery was also used to examine the role of ANGPTL8 in mice. RESULTS: ANGPTL8 is significantly up-regulated in amino acid-deprived cultured cells in vitro. The activation of ANGPTL8 gene transcription was mediated through the RAS/c-RAF/MAPK signaling pathway rather than the general GCN2/ATF4 pathways. ANGPTL8 activated the ERK signal transduction pathway in hepatocytes, adipocytes, and pancreatic ß-cells, up-regulating early growth response transcription factor (Egr1) and down-regulating adipose triglyceride lipase (ATGL). CONCLUSION: ANGPTL8 is a stress-response protein that regulates fat metabolism by suppressing ATGL expression, revealing a mechanistic connection between ANGPTL8 and lipid homeostasis in mammalian cells.


Subject(s)
Adipocytes/metabolism , Angiopoietins/genetics , Lipase/genetics , Triglycerides/metabolism , 3T3-L1 Cells , Activating Transcription Factor 4/genetics , Activating Transcription Factor 4/metabolism , Adipocytes/cytology , Adipocytes/drug effects , Angiopoietin-Like Protein 8 , Angiopoietin-like Proteins , Angiopoietins/metabolism , Angiopoietins/pharmacology , Animals , Cell Differentiation , Cell Line , Connective Tissue Growth Factor/genetics , Connective Tissue Growth Factor/metabolism , Culture Media/chemistry , Culture Media/pharmacology , Early Growth Response Protein 1/genetics , Early Growth Response Protein 1/metabolism , Gene Expression Regulation , Glycerol/metabolism , Hep G2 Cells , Humans , Insulin-Secreting Cells/cytology , Insulin-Secreting Cells/drug effects , Insulin-Secreting Cells/metabolism , Lipase/metabolism , Lipid Metabolism/drug effects , Male , Mice , Mice, Inbred ICR , Mitogen-Activated Protein Kinases/genetics , Mitogen-Activated Protein Kinases/metabolism , Proto-Oncogene Proteins c-raf/genetics , Proto-Oncogene Proteins c-raf/metabolism , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Recombinant Proteins/pharmacology , Signal Transduction , ras Proteins/genetics , ras Proteins/metabolism
11.
PLoS One ; 10(7): e0131176, 2015.
Article in English | MEDLINE | ID: mdl-26132105

ABSTRACT

OBJECTIVES: Angiopoietin-like protein 2 (ANGPTL2), a recently identified pro-inflammatory cytokine, is mainly secreted from the adipose tissue. This study aimed to explore the role of ANGPTL2 in adipose tissue inflammation and macrophage activation in a mouse model of diabetes. METHODOLOGY/PRINCIPAL FINDINGS: Adenovirus mediated lacZ (Ad-LacZ) or human ANGPTL2 (Ad-ANGPTL2) was delivered via tail vein in diabetic db/db mice. Ad-ANGPTL2 treatment for 2 weeks impaired both glucose tolerance and insulin sensitivity as compared to Ad-LacZ treatment. Ad-ANGPTL2 treatment significantly induced pro-inflammatory gene expression in white adipose tissue. We also isolated stromal vascular fraction from epididymal fat pad and analyzed adipose tissue macrophage and T lymphocyte populations by flow cytometry. Ad-ANGPTL2 treated mice had more adipose tissue macrophages (F4/80+CD11b+) and a larger M1 macrophage subpopulation (F4/80+CD11b+CD11c+). Moreover, Ad-ANGPTL2 treatment increased a CD8-positive T cell population in adipose tissue, which preceded increased macrophage accumulation. Consistent with our in vivo results, recombinant human ANGPTL2 protein treatment increased mRNA levels of pro-inflammatory gene products and production of TNF-α protein in the human macrophage-like cell line THP-1. Furthermore, Ad-ANGPTL2 treatment induced lipid accumulation and increased fatty acid synthesis, lipid metabolism related gene expression in mouse liver. CONCLUSION: ANGPTL2 treatment promotes macrophage accumulation and activation. These results suggest potential mechanisms for insulin resistance.


Subject(s)
Adipose Tissue, White/metabolism , Angiopoietins/metabolism , Diabetes Mellitus, Experimental/metabolism , Macrophages/metabolism , Obesity/metabolism , T-Lymphocytes/metabolism , Adenoviridae/genetics , Adipose Tissue, White/drug effects , Adipose Tissue, White/pathology , Angiopoietin-Like Protein 2 , Angiopoietin-like Proteins , Angiopoietins/genetics , Angiopoietins/pharmacology , Animals , Antigens, Differentiation/genetics , Antigens, Differentiation/metabolism , CD11b Antigen/genetics , CD11b Antigen/metabolism , CD11c Antigen/genetics , CD11c Antigen/metabolism , Cell Movement/drug effects , Diabetes Mellitus, Experimental/genetics , Diabetes Mellitus, Experimental/pathology , Genetic Vectors , Glucose Tolerance Test , Humans , Insulin Resistance , Lipid Metabolism/drug effects , Liver/drug effects , Liver/metabolism , Liver/pathology , Macrophages/drug effects , Macrophages/pathology , Male , Mice , Mice, Inbred C57BL , Obesity/genetics , Obesity/pathology , Signal Transduction , T-Lymphocytes/drug effects , T-Lymphocytes/pathology , Tumor Necrosis Factor-alpha/biosynthesis , Tumor Necrosis Factor-alpha/genetics
12.
Stem Cells Transl Med ; 4(9): 1064-72, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26160960

ABSTRACT

UNLABELLED: Human cord blood (CB)-derived hematopoietic stem cells (HSCs) are an interesting source for HSC transplantation. However, the number of collected CB-HSCs is often too low for one transplantation; therefore, ex vivo expansion of CB-HSCs is desirable. Current expansion protocols are based on the use of cytokine combinations, including insulin-like growth factor-binding protein 2 (IGFBP2) and angiopoietin-like proteins, or combinations with "small molecules" such as stemregenin-1. The aim of our project was to compare the potential of different CB-HSC expansion strategies side-by-side by phenotypical analysis in vitro and serial engraftment properties in NOD/SCID/IL2rg-/- (NSG) immunodeficient mice. We further identified resveratrol, a naturally occurring polyphenol, as a new, alternative small molecule combined with cytokines to facilitate serum-free ex vivo expansion of human CB-HSCs. The cultivation in resveratrol preserved the CB-HSC phenotype in vitro most efficiently and was ∼2 times more potent than commonly used cytokine conditions (including stem cell factor, thrombopoietin, Fms-related tyrosine kinase 3 ligand, interleukin-6) and the recently established serum-free culture, including IGFBP2 and angiopoietin-like 5. Serial transplantation studies further confirmed resveratrol to support robust multilineage engraftment in primary and secondary NSG recipients. Therefore, our work proposes resveratrol as a new small molecule for improved ex vivo culture and modification of human HSCs based on an efficient ex vivo propagation of the HSC fate. SIGNIFICANCE: Human cord blood (CB)-derived hematopoietic stem cells (HSCs) are an important source for HSC transplantations but restricted in their usage because of their low numbers. In gene therapy, modifications of HSCs relies on their ex vivo modification without losing their stemness properties. Therefore, ex vivo cultivation and expansion of CB-HSCs is important for their effective application in HSC transplantation and gene therapy. Several promising protocols for serum-free cultivation of HSCs using different combinations of cytokines or so-called small molecules are described. A direct comparison was performed of three described serum-free cytokine conditions, demonstrating that the natural occurring polyphenol resveratrol is able to support ex vivo cultivation of CB-HSCs. The results show that resveratrol is an additional candidate for improving ex vivo cultures of HSCs for transplantation and gene therapeutic applications in the future.


Subject(s)
Antioxidants/pharmacology , Fetal Blood/drug effects , Hematopoietic Stem Cell Transplantation , Hematopoietic Stem Cells/drug effects , Stilbenes/pharmacology , Angiopoietins/pharmacology , Animals , Fetal Blood/cytology , Fetal Blood/metabolism , Graft Survival , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism , Humans , Insulin-Like Growth Factor Binding Protein 2/pharmacology , Interleukin Receptor Common gamma Subunit/deficiency , Interleukin Receptor Common gamma Subunit/genetics , Interleukin-6/pharmacology , Mice , Mice, Inbred NOD , Mice, Knockout , Mice, SCID , Primary Cell Culture , Resveratrol , Stem Cell Factor/pharmacology , Thrombopoietin/pharmacology , Transplantation, Heterologous , Vascular Endothelial Growth Factor Receptor-1/pharmacology
13.
Int J Oncol ; 46(4): 1651-8, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25651778

ABSTRACT

Angiopoietin-like protein 2 (ANGPTL2) plays an important role in inflammatory carcinogenesis and tumor metastasis. The compound GDC-0152 is a peptidomimetic small molecule antagonist of inhibitor of apoptosis (IAP) proteins with antitumor activity. However, the interaction between ANGPTL2 and GDC-0152 has not been studied. It has been proven that ANGPTL2 promotes metastasis of osteosarcoma. Therefore, in the present study, the effect of GDC-0152 on the malignant progression of osteosarcoma promoted by ANGPTL2 was investigated. Human osteosarcoma cell line SaOS2 cells were pre-treated or non-treated with GDC-0152 and then exposed to recombinant human ANGPTL2. The viability of SaOS2 cells was determined by MTT assay, the migration of SaOS2 cells was analyzed by chamber migration assay kit, and the SaOS2 cell apoptosis was determined by fluorescence-activated cell sorting (FACS) and nuclear staining. Treatment with ANGPTL2 increased SaOS2 cell growth and migration and decreased cell apoptosis. The increased cell growth and decreased cell apoptosis were significantly attenuated in SaOS2 cells receiving GDC-0152. However, the ANGPTL2-increased SaOS2 cell migration was not inhibited by GDC-0152 treatment. Furthermore, western blot analysis showed that the activation of phosphatidyl inositol 3-kinase (PI3K) (p85), PI3K (p110), protein kinase B (Akt) (Ser473), Akt (Thr308) and p38 mitogen-activated protein kinase (p38MAPK) were upregulated by ANGPTL2. Quantitative real-time polymerase chain reaction (qTR-PCR) and gelatin zymography showed that the mRNA expression and activity of matrix metalloproteinase-9 (MMP-9) and matrix metalloproteinase-2 (MMP-2) were also increased by ANGPTL2. The upregulated activation of PI3K and Akt were significantly suppressed by the treatment of GDC-0152. In contrast, GDC-0152 did not suppress ANGPTL2-induced p38MAPK phosphorylation, MMP-9/MMP-2 mRNA expression or MMP-9/MMP-2 activity. Taken together, these data indicate that GDC-0152 attenuates the malignant progression of osteosarcoma promoted by ANGPTL2 via PI3K/AKT but not p38MAPK signaling pathway. The present study indicated a novel therapeutic strategy to inhibit tumor growth by indirectly preventing ANGPTL2 signaling.


Subject(s)
Antineoplastic Agents/pharmacology , Bone Neoplasms/drug therapy , Cyclohexanes/pharmacology , MAP Kinase Signaling System/drug effects , Osteosarcoma/drug therapy , Pyrroles/pharmacology , Angiopoietin-Like Protein 2 , Angiopoietin-like Proteins , Angiopoietins/pharmacology , Apoptosis/drug effects , Bone Neoplasms/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , Humans , Osteosarcoma/pathology , Recombinant Proteins/pharmacology , Up-Regulation
14.
Haematologica ; 100(5): 585-94, 2015 May.
Article in English | MEDLINE | ID: mdl-25637050

ABSTRACT

Successful expansion of hematopoietic stem cells would benefit the use of hematopoietic stem cell transplants in the clinic. Several angiopoietin-like proteins, including angiopoietin-like 7, can support the activity of hematopoietic stem cells. However, effects of ANGPTL7 on human hematopoietic stem cells and the downstream signaling cascade activated by ANGPTL7 are poorly understood. Here, we established a human hematopoietic stem and progenitor cell-supportive mouse fetal liver cell line that specifically expressed the Angptl7 protein. Furthermore, we found ANGPTL7 is capable of stimulating human hematopoietic stem and progenitor cell expansion and increasing the repopulation activities of human hematopoietic progenitors in xenografts. RNA-sequencing analysis showed that ANGPTL7 activated the expression of CXCR4, HOXB4 and Wnt downstream targets in human hematopoietic progenitors. In addition, chemical manipulation of Wnt signaling diminished the effects of ANGPTL7 on human hematopoietic stem and progenitor cells in culture. In summary, we identify the secreted growth factor ANGPTL7 as a regulator of both human hematopoietic stem and progenitor cell expansion and regeneration.


Subject(s)
Angiopoietins/genetics , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism , Angiopoietin-Like Protein 7 , Angiopoietin-like Proteins , Angiopoietins/pharmacology , Animals , Cell Communication , Cluster Analysis , Coculture Techniques , Fetal Blood/cytology , Gene Expression , Gene Expression Profiling , Hematopoietic Stem Cells/drug effects , Humans , Mice , Mice, Knockout , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/pharmacology , Stromal Cells/metabolism , Wnt Signaling Pathway/drug effects
15.
J Cell Biochem ; 116(1): 45-57, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25145474

ABSTRACT

A key feature in the induction of pathological angiogenesis is that inflammation precedes and accompanies the formation of neovessels as evidenced by increased vascular permeability and the recruitment of inflammatory cells. Previously, we and other groups have shown that selected growth factors, namely vascular endothelial growth factor (VEGF) and angiopoietins (Ang1 and Ang2) do not only promote angiogenesis, but can also induce inflammatory response. Herein, given a pro-inflammatory environment, we addressed the individual capacity of VEGF and angiopoietins to promote the formation of mature neovessels and to identify the different types of inflammatory cells accompanying the angiogenic process over time. Sterilized polyvinyl alcohol (PVA) sponges soaked in growth factor-depleted Matrigel mixed with PBS, VEGF, Ang1, or Ang2 (200 ng/200 µl) were subcutaneously inserted into anesthetized mice. Sponges were removed at day 4, 7, 14, or 21 post-procedure for histological, immunohistological (IHC), and flow cytometry analyses. As compared to PBS-treated sponges, the three growth factors promoted the recruitment of inflammatory cells, mainly neutrophils and macrophages, and to a lesser extent, T- and B-cells. In addition, they were more potent and more rapid in the recruitment of endothelial cells (ECs) and in the formation and maturation (ensheating of smooth muscle cells around ECs) of neovessels. Thus, the autocrine/paracrine interaction among the different inflammatory cells in combination with VEGF, Ang1, or Ang2 provides a suitable microenvironment for the formation and maturation of blood vessels.


Subject(s)
Angiopoietins/pharmacology , Vascular Endothelial Growth Factor A/pharmacology , Angiopoietin-1/pharmacology , Angiopoietin-2/pharmacology , Animals , Collagen/chemistry , Drug Combinations , Flow Cytometry , Immunohistochemistry , Laminin/chemistry , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Neovascularization, Physiologic/drug effects , Phagocytosis/genetics , Phagocytosis/physiology , Proteoglycans/chemistry
16.
Transl Res ; 165(4): 499-504, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25005737

ABSTRACT

The development of new and specific treatment options for kidney disease in general and glomerular diseases in specific has lagged behind other fields like heart disease and cancer. As a result, nephrologists have had to test and adapt therapeutics developed for other indications to treat glomerular diseases. One of the major factors contributing to this inertia has been the poor understanding of disease mechanisms. One way to elucidate these disease mechanisms is to study the association between the cardinal manifestations of glomerular diseases. Because many of these patients develop nephrotic syndrome, understanding the relationship of proteinuria, the primary driver in this syndrome, with hypoalbuminemia, hypercholesterolemia, hypertriglyceridemia, edema, and lipiduria could provide valuable insight. The recent unraveling of the relationship between proteinuria and hypertriglyceridemia mediated by free fatty acids, albumin, and the secreted glycoprotein angiopoietin-like 4 (Angptl4) offers a unique opportunity to develop novel therapeutics for glomerular diseases. In this review, the therapeutic potential of mutant forms of Angptl4 in reducing proteinuria and, as a consequence, alleviating the other manifestations of nephrotic syndrome is discussed.


Subject(s)
Hypertriglyceridemia/complications , Nephrotic Syndrome/drug therapy , Proteinuria/complications , Angiopoietin-Like Protein 4 , Angiopoietins/genetics , Angiopoietins/pharmacology , Humans , Nephrotic Syndrome/etiology , Recombinant Proteins
17.
Diabetes ; 64(4): 1142-53, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25338813

ABSTRACT

Hypothalamic lipid sensing is important for the maintenance of energy balance. Angiopoietin-like protein 3 (Angptl3) critically regulates the clearance of circulating lipids by inhibiting lipoprotein lipase (LPL). The current study demonstrated that Angptl3 is highly expressed in the neurons of the mediobasal hypothalamus, an important area in brain lipid sensing. Suppression of hypothalamic Angptl3 increased food intake but reduced energy expenditure and fat oxidation, thereby promoting weight gain. Consistently, intracerebroventricular (ICV) administration of Angptl3 caused the opposite metabolic changes, supporting an important role for hypothalamic Angptl3 in the control of energy balance. Notably, ICV Angptl3 significantly stimulated hypothalamic LPL activity. Moreover, coadministration of the LPL inhibitor apolipoprotein C3 antagonized the effects of Angptl3 on energy metabolism, indicating that LPL activation is critical for the central metabolic actions of Angptl3. Increased LPL activity is expected to promote lipid uptake by hypothalamic neurons, leading to enhanced brain lipid sensing. Indeed, ICV injection of Angptl3 increased long-chain fatty acid (LCFA) and LCFA-CoA levels in the hypothalamus. Furthermore, inhibitors of hypothalamic lipid-sensing pathways prevented Angptl3-induced anorexia and weight loss. These findings identify Angptl3 as a novel regulator of the hypothalamic lipid-sensing pathway.


Subject(s)
Angiopoietins/metabolism , Energy Metabolism/physiology , Fatty Acids/metabolism , Hypothalamus/metabolism , Lipoprotein Lipase/metabolism , Angiopoietin-Like Protein 3 , Angiopoietin-like Proteins , Angiopoietins/genetics , Angiopoietins/pharmacology , Animals , Avoidance Learning/drug effects , Avoidance Learning/physiology , Conditioning, Operant/drug effects , Conditioning, Operant/physiology , Eating/drug effects , Eating/physiology , Energy Metabolism/drug effects , Hypothalamus/drug effects , Male , Mice , RNA Interference , Rats , Rats, Sprague-Dawley
18.
PLoS One ; 9(8): e105642, 2014.
Article in English | MEDLINE | ID: mdl-25170927

ABSTRACT

Allogeneic hematopoietic stem cell (HSC) transplantations from umbilical cord blood or autologous HSCs for gene therapy purposes are hampered by limited number of stem cells. To test the ability to expand HSCs in vitro prior to transplantation, two growth factor cocktails containing stem cell factor, thrombopoietin, fms-related tyrosine kinase-3 ligand (STF) or stem cell factor, thrombopoietin, insulin-like growth factor-2, fibroblast growth factor-1 (STIF) either with or without the addition of angiopoietin-like protein-3 (Angptl3) were used. Culturing HSCs in STF and STIF media for 7 days expanded long-term repopulating stem cells content in vivo by ∼6-fold and ∼10-fold compared to freshly isolated stem cells. Addition of Angptl3 resulted in increased expansion of these populations by ∼17-fold and ∼32-fold, respectively, and was further supported by enforced expression of Angptl3 in HSCs through lentiviral transduction that also promoted HSC expansion. As expansion of highly purified lineage-negative, Sca-1+, c-Kit+ HSCs was less efficient than less pure lineage-negative HSCs, Angptl3 may have a direct effect on HCS but also an indirect effect on accessory cells that support HSC expansion. No evidence for leukemia or toxicity was found during long-term follow up of mice transplanted with ex vivo expanded HSCs or manipulated HSC populations that expressed Angptl3. We conclude that the cytokine combinations used in this study to expand HSCs ex vivo enhances the engraftment in vivo. This has important implications for allogeneic umbilical cord-blood derived HSC transplantations and autologous HSC applications including gene therapy.


Subject(s)
Angiopoietins/genetics , Cell Proliferation/genetics , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism , Angiopoietin-Like Protein 3 , Angiopoietin-like Proteins , Angiopoietins/metabolism , Angiopoietins/pharmacology , Animals , Antigens, Ly/metabolism , Cell Proliferation/drug effects , Cells, Cultured , Female , Fibroblast Growth Factor 1/pharmacology , Flow Cytometry , Genetic Vectors/genetics , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , HEK293 Cells , HeLa Cells , Hematopoietic Stem Cell Transplantation/methods , Hematopoietic Stem Cells/drug effects , Humans , Insulin-Like Growth Factor II/pharmacology , Lentivirus/genetics , Male , Membrane Proteins/metabolism , Mice, Inbred BALB C , Proto-Oncogene Proteins c-kit/metabolism , Stem Cell Factor/pharmacology , Thrombopoietin/pharmacology , Transfection
20.
Mol Ther ; 22(9): 1593-604, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24903577

ABSTRACT

Impaired wound healing is a major source of morbidity in diabetic patients. Poor outcome has, in part, been related to increased inflammation, poor angiogenesis, and deficiencies in extracellular matrix components. Despite the enormous impact of these chronic wounds, effective therapies are lacking. Here, we showed that the topical application of recombinant matricellular protein angiopoietin-like 4 (ANGPTL4) accelerated wound reepithelialization in diabetic mice, in part, by improving angiogenesis. ANGPTL4 expression is markedly elevated upon normal wound injury. In contrast, ANGPTL4 expression remains low throughout the healing period in diabetic wounds. Exogenous ANGPTL4 modulated several regulatory networks involved in cell migration, angiogenesis, and inflammation, as evidenced by an altered gene expression signature. ANGPTL4 influenced the expression profile of endothelial-specific CD31 in diabetic wounds, returning its profile to that observed in wild-type wounds. We showed ANGPTL4-induced nitric oxide production through an integrin/JAK/STAT3-mediated upregulation of inducible nitric oxide synthase (iNOS) expression in wound epithelia, thus revealing a hitherto unknown mechanism by which ANGPTL4 regulated angiogenesis via keratinocyte-to-endothelial-cell communication. These data show that the replacement of ANGPTL4 may be an effective adjunctive or new therapeutic avenue for treating poor healing wounds. The present finding also confirms that therapeutic angiogenesis remains an attractive treatment modality for diabetic wound healing.


Subject(s)
Angiopoietins/administration & dosage , Diabetes Mellitus, Experimental/complications , Neovascularization, Physiologic/drug effects , Nitric Oxide Synthase Type II/metabolism , STAT3 Transcription Factor/metabolism , Angiopoietins/pharmacology , Animals , Cell Communication , Diabetes Mellitus, Experimental/genetics , Diabetes Mellitus, Experimental/metabolism , Gene Expression Regulation/drug effects , Keratinocytes/metabolism , Mice , Re-Epithelialization , Recombinant Proteins/genetics , Recombinant Proteins/pharmacology , Signal Transduction
SELECTION OF CITATIONS
SEARCH DETAIL
...