1
|
Patidar K, Versypt ANF. Logic-Based Modeling of Inflammatory Macrophage Crosstalk with Glomerular Endothelial Cells in Diabetic Kidney Disease. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.04.04.535594. [PMID: 37066138 PMCID: PMC10104015 DOI: 10.1101/2023.04.04.535594] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/19/2023]
Abstract
Diabetic kidney disease is a complication in one out of three patients with diabetes. Aberrant glucose metabolism in diabetes leads to structural and functional damage in glomerular tissue and a systemic inflammatory immune response. Complex cellular signaling is at the core of metabolic and functional derangement. Unfortunately, the mechanism underlying the role of inflammation in glomerular endothelial cell dysfunction during diabetic kidney disease is not fully understood. Mathematical models in systems biology allow the integration of experimental evidence and cellular signaling networks to understand mechanisms involved in disease progression. This study developed a logic-based ordinary differential equations model to study inflammatory crosstalk between macrophages and glomerular endothelial cells during diabetic kidney disease progression using a protein signaling network stimulated with glucose and lipopolysaccharide. This modeling approach reduced the biological parameters needed to study signaling networks. The model was fitted to and validated against available biochemical data from \textit{in vitro} experiments. The model identified mechanisms for dysregulated signaling in macrophages and glomerular endothelial cells during diabetic kidney disease. In addition, the influence of signaling interactions on glomerular endothelial cell morphology through selective knockdown and downregulation was investigated. Simulation results showed that partial knockdown of VEGF receptor 1, PLC-γ, adherens junction proteins, and calcium partially recovered the intercellular gap width between glomerular endothelial cells. These findings contribute to understanding signaling and molecular perturbations that affect the glomerular endothelial cells in the early stage of diabetic kidney disease.
Collapse
|
2
|
Hu SY, Xue CD, Li YJ, Li S, Gao ZN, Qin KR. Microfluidic investigation for shear-stress-mediated repair of dysglycemia-induced endothelial cell damage. MECHANOBIOLOGY IN MEDICINE 2024; 2:100069. [DOI: 10.1016/j.mbm.2024.100069] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
3
|
Shi Z, Yao C, Shui Y, Li S, Yan H. Research progress on the mechanism of angiogenesis in wound repair and regeneration. Front Physiol 2023; 14:1284981. [PMID: 38089479 PMCID: PMC10711283 DOI: 10.3389/fphys.2023.1284981] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Accepted: 11/13/2023] [Indexed: 12/10/2024] Open
Abstract
Poor wound healing and pathological healing have been pressing issues in recent years, as they impact human quality of life and pose risks of long-term complications. The study of neovascularization has emerged as a prominent research focus to address these problems. During the process of repair and regeneration, the establishment of a new vascular system is an indispensable stage for complete healing. It provides favorable conditions for nutrient delivery, oxygen supply, and creates an inflammatory environment. Moreover, it is a key manifestation of the proliferative phase of wound healing, bridging the inflammatory and remodeling phases. These three stages are closely interconnected and inseparable. This paper comprehensively integrates the regulatory mechanisms of new blood vessel formation in wound healing, focusing on the proliferation and migration of endothelial cells and the release of angiogenesis-related factors under different healing outcomes. Additionally, the hidden link between the inflammatory environment and angiogenesis in wound healing is explored.
Collapse
Affiliation(s)
- Zhuojun Shi
- Department of Plastic and Burns Surgery, The Affiliated Hospital of Southwest Medical University, National Key Clinical Construction Specialty, Wound Repair and Regeneration Laboratory, Luzhou, Sichuan, China
| | - Chong Yao
- Department of Plastic and Burns Surgery, The Affiliated Hospital of Southwest Medical University, National Key Clinical Construction Specialty, Wound Repair and Regeneration Laboratory, Luzhou, Sichuan, China
| | - Yujie Shui
- Department of Plastic and Burns Surgery, The Affiliated Hospital of Southwest Medical University, National Key Clinical Construction Specialty, Wound Repair and Regeneration Laboratory, Luzhou, Sichuan, China
| | - Site Li
- Department of Plastic and Burns Surgery, The Affiliated Hospital of Southwest Medical University, National Key Clinical Construction Specialty, Wound Repair and Regeneration Laboratory, Luzhou, Sichuan, China
| | - Hong Yan
- Laboratory of Plastic Surgery, Department of Plastic Surgery and Reconstruction, Second Hospital of West China, Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
4
|
Lin LQ, Zeng HK, Luo YL, Chen DF, Ma XQ, Chen HJ, Song XY, Wu HK, Li SY. Mechanical stretch promotes apoptosis and impedes ciliogenesis of primary human airway basal stem cells. Respir Res 2023; 24:237. [PMID: 37773064 PMCID: PMC10540374 DOI: 10.1186/s12931-023-02528-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Accepted: 08/31/2023] [Indexed: 09/30/2023] Open
Abstract
BACKGROUND Airway basal stem cells (ABSCs) have self-renewal and differentiation abilities. Although an abnormal mechanical environment related to chronic airway disease (CAD) can cause ABSC dysfunction, it remains unclear how mechanical stretch regulates the behavior and structure of ABSCs. Here, we explored the effect of mechanical stretch on primary human ABSCs. METHODS Primary human ABSCs were isolated from healthy volunteers. A Flexcell FX-5000 Tension system was used to mimic the pathological airway mechanical stretch conditions of patients with CAD. ABSCs were stretched for 12, 24, or 48 h with 20% elongation. We first performed bulk RNA sequencing to identify the most predominantly changed genes and pathways. Next, apoptosis of stretched ABSCs was detected with Annexin V-FITC/PI staining and a caspase 3 activity assay. Proliferation of stretched ABSCs was assessed by measuring MKI67 mRNA expression and cell cycle dynamics. Immunofluorescence and hematoxylin-eosin staining were used to demonstrate the differentiation state of ABSCs at the air-liquid interface. RESULTS Compared with unstretched control cells, apoptosis and caspase 3 activation of ABSCs stretched for 48 h were significantly increased (p < 0.0001; p < 0.0001, respectively), and MKI67 mRNA levels were decreased (p < 0.0001). In addition, a significant increase in the G0/G1 population (20.2%, p < 0.001) and a significant decrease in S-phase cells (21.1%, p < 0.0001) were observed. The ratio of Krt5+ ABSCs was significantly higher (32.38% vs. 48.71%, p = 0.0037) following stretching, while the ratio of Ac-tub+ cells was significantly lower (37.64% vs. 21.29%, p < 0.001). Moreover, compared with the control, the expression of NKX2-1 was upregulated significantly after stretching (14.06% vs. 39.51%, p < 0.0001). RNA sequencing showed 285 differentially expressed genes, among which 140 were upregulated and 145 were downregulated, revealing that DDIAS, BIRC5, TGFBI, and NKX2-1 may be involved in the function of primary human ABSCs during mechanical stretch. There was no apparent difference between stretching ABSCs for 24 and 48 h compared with the control. CONCLUSIONS Pathological stretching induces apoptosis of ABSCs, inhibits their proliferation, and disrupts cilia cell differentiation. These features may be related to abnormal regeneration and repair observed after airway epithelium injury in patients with CAD.
Collapse
Affiliation(s)
- Li-Qin Lin
- The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, Guangdong, China
- National Clinical Research Center for Respiratory Disease, Guangzhou, 510120, Guangdong, China
- Guangzhou Institute of Respiratory Health, Guangzhou, 510120, Guangdong, China
- State Key Laboratory of Respiratory Disease, Guangzhou, 511495, Guangdong, China
| | - Hai-Kang Zeng
- The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, Guangdong, China
- National Clinical Research Center for Respiratory Disease, Guangzhou, 510120, Guangdong, China
- Guangzhou Institute of Respiratory Health, Guangzhou, 510120, Guangdong, China
- State Key Laboratory of Respiratory Disease, Guangzhou, 511495, Guangdong, China
| | - Yu-Long Luo
- The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510799, Guangdong, China
- Key Laboratory of Biological Targeting Diagnosis, Guangzhou, 510799, Guangdong, China
- Therapy and Rehabilitation of Guangdong Higher Education Institutes, Guangzhou, 510799, Guangdong, China
- Innovation Centre for Advanced Interdisciplinary Medicine, Guangzhou, 510799, Guangdong, China
| | - Di-Fei Chen
- The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, Guangdong, China
- National Clinical Research Center for Respiratory Disease, Guangzhou, 510120, Guangdong, China
- Guangzhou Institute of Respiratory Health, Guangzhou, 510120, Guangdong, China
- State Key Laboratory of Respiratory Disease, Guangzhou, 511495, Guangdong, China
| | - Xiao-Qian Ma
- The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, Guangdong, China
- National Clinical Research Center for Respiratory Disease, Guangzhou, 510120, Guangdong, China
- Guangzhou Institute of Respiratory Health, Guangzhou, 510120, Guangdong, China
- State Key Laboratory of Respiratory Disease, Guangzhou, 511495, Guangdong, China
| | - Huan-Jie Chen
- The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, Guangdong, China
- National Clinical Research Center for Respiratory Disease, Guangzhou, 510120, Guangdong, China
- Guangzhou Institute of Respiratory Health, Guangzhou, 510120, Guangdong, China
- State Key Laboratory of Respiratory Disease, Guangzhou, 511495, Guangdong, China
| | - Xin-Yu Song
- The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, Guangdong, China
- National Clinical Research Center for Respiratory Disease, Guangzhou, 510120, Guangdong, China
- Guangzhou Institute of Respiratory Health, Guangzhou, 510120, Guangdong, China
- State Key Laboratory of Respiratory Disease, Guangzhou, 511495, Guangdong, China
| | - Hong-Kai Wu
- The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, Guangdong, China
- National Clinical Research Center for Respiratory Disease, Guangzhou, 510120, Guangdong, China
- Guangzhou Institute of Respiratory Health, Guangzhou, 510120, Guangdong, China
- State Key Laboratory of Respiratory Disease, Guangzhou, 511495, Guangdong, China
| | - Shi-Yue Li
- The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, Guangdong, China.
- National Clinical Research Center for Respiratory Disease, Guangzhou, 510120, Guangdong, China.
- Guangzhou Institute of Respiratory Health, Guangzhou, 510120, Guangdong, China.
- State Key Laboratory of Respiratory Disease, Guangzhou, 511495, Guangdong, China.
| |
Collapse
|
5
|
Sunartvanichkul T, Arayapisit T, Sangkhamanee SS, Chaweewannakorn C, Iwasaki K, Klaihmon P, Sritanaudomchai H. Stem cell-derived exosomes from human exfoliated deciduous teeth promote angiogenesis in hyperglycemic-induced human umbilical vein endothelial cells. J Appl Oral Sci 2023; 31:e20220427. [PMID: 37042872 PMCID: PMC10118382 DOI: 10.1590/1678-7757-2022-0427] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Revised: 01/12/2023] [Accepted: 02/07/2023] [Indexed: 04/13/2023] Open
Abstract
OBJECTIVE To investigate the angiogenesis in human umbilical vein endothelial cells (HUVEC) under high glucose concentration, treated with exosomes derived from stem cells from human exfoliated deciduous teeth (SHED). METHODOLOGY SHED-derived exosomes were isolated by differential centrifugation and were characterized by nanoparticle tracking analysis, transmission electron microscopy, and flow cytometric assays. We conducted in vitro experiments to examine the angiogenesis in HUVEC under high glucose concentration. Cell Counting Kit-8, migration assay, tube formation assay, quantitative real-time PCR, and immunostaining were performed to study the role of SHED-derived exosomes in cell proliferation, migration, and angiogenic activities. RESULTS The characterization confirmed SHED-derived exosomes: size ranged from 60-150 nm with a mode of 134 nm, cup-shaped morphology, and stained positively for CD9, CD63, and CD81. SHED-exosome significantly enhanced the proliferation and migration of high glucose-treated HUVEC. A significant reduction was observed in tube formation and a weak CD31 staining compared to the untreated-hyperglycemic-induced group. Interestingly, exosome treatment improved tube formation qualitatively and demonstrated a significant increase in tube formation in the covered area, total branching points, total tube length, and total loop parameters. Moreover, SHED-exosome upregulates angiogenesis-related factors, including the GATA2 gene and CD31 protein. CONCLUSIONS Our data suggest that the use of SHED-derived exosomes potentially increases angiogenesis in HUVEC under hyperglycemic conditions, which includes increased cell proliferation, migration, tubular structures formation, GATA2 gene, and CD31 protein expression. SHED-exosome usage may provide a new treatment strategy for periodontal patients with diabetes mellitus.
Collapse
Affiliation(s)
| | - Tawepong Arayapisit
- Mahidol University, Faculty of Dentistry, Department of Anatomy, Bangkok, Thailand
| | | | | | - Kengo Iwasaki
- Osaka Dental University, Advanced Medical Research Center, Translational Research Institute for Medical Innovation, Osaka, Japan
| | - Phatchanat Klaihmon
- Mahidol University, Faculty of Medicine Siriraj Hospital, Siriraj Center of Excellence for Stem Cell Research, Bangkok, Thailand
| | | |
Collapse
|
6
|
Hydroquinone predisposes for retinal pigment epithelial (RPE) cell degeneration in inflammatory conditions. Immunol Res 2022; 70:678-687. [PMID: 35661979 PMCID: PMC9499922 DOI: 10.1007/s12026-022-09300-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Accepted: 05/24/2022] [Indexed: 12/14/2022]
Abstract
In addition to hypoxia, inflammation is capable of inducing vascular endothelial growth factor (VEGF) expression in human retinal pigment epithelial (RPE) cells. Excessive levels of VEGF promote choroidal neovascularization and thereby contribute to the pathogenesis of wet age-related macular degeneration (AMD). Intravitreal anti-VEGF injections ameliorate pathological vessel neoformation in wet AMD but excessive dampening of VEGF can result in a degeneration of the RPE. In the present study, we induced VEGF production by exposing human ARPE-19 cells to the pro-inflammatory IL-1α and subsequently to hydroquinone, a component of tobacco smoke that is a major environmental risk factor for AMD. Effects were monitored by measuring the levels of VEGF and anti-angiogenic pigment epithelium-derived factor (PEDF) using an enzyme-linked immunosorbent assay (ELISA) technique. In addition, we measured the production of reactive oxygen species (ROS) using the 2′,7′-dichlorofluorescin diacetate (H2DCFDA) probe and studied the effects of two anti-oxidants, ammonium pyrrolidinedithiocarbamate (APDC) and N-acetyl-cysteine (NAC), on VEGF production. Cellular and secreted VEGF as well as secreted PEDF levels were reduced at all tested hydroquinone concentrations (10, 50, or 200 µM); these effects were evident prior to any reduction of cell viability evoked by hydroquinone. Cell viability was carefully explored in our previous study and verified by microscoping in the present study. APDC further reduced the VEGF levels, whereas NAC increased them. The 50 μM concentration of hydroquinone increased ROS production in ARPE-19 cells primed with IL-1α. Hydroquinone disturbs the regulatory balance of VEGF and PEDF in inflammatory conditions. These data support the idea that hydroquinone mediates RPE degeneration by reducing VEGF levels and may predispose to dry AMD since VEGF is as well important for retinal integrity.
Collapse
|
7
|
Myricitrin exerts protective effect on retina in diabetic retinopathy via modulating oxidative stress expression of VEGF and apoptosis in experimental rats: a docking confirmation study. Mol Cell Toxicol 2022. [DOI: 10.1007/s13273-021-00167-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
|
8
|
Ciechanowska A, Gora I, Sabalinska S, Foltynski P, Ladyzynski P. Effect of glucose concentration and culture substrate on HUVECs viability in in vitro cultures: A literature review and own results. Biocybern Biomed Eng 2021. [DOI: 10.1016/j.bbe.2021.04.010] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
9
|
Gastaldi G, Pannier F, Roztočil K, Lugli M, Mansilha A, Haller H, Rabe E, VAN Rijn MJ. Chronic venous disease and diabetic microangiopathy: pathophysiology and commonalities. INT ANGIOL 2021; 40:457-469. [PMID: 34547884 DOI: 10.23736/s0392-9590.21.04664-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Chronic venous disease and diabetes mellitus are highly prevalent and debilitating conditions affecting millions of individuals globally. Although these conditions are typically considered as separate entities, they often co-exist which may be important in both understanding their pathophysiology and determining the best treatment strategy. Diabetes mellitus is twice as common in patients with chronic venous disease compared with the general population. Notably, a large proportion of patients with diabetes mellitus present with venous disorders, although this is often overlooked. The etiology of chronic venous disease is multifactorial, involving hemodynamic, genetic, and environmental factors which result in changes to the venous endothelium and structural wall as well as inflammation. Inflammation, endothelial dysfunction and hyperfiltration or leakage, are commonly observed in diabetes mellitus and cause various diabetic microvascular complications. Both diseases are also influenced by the increased expression of adhesion molecules, chemokines, and cytokines, and are characterized by the presence of vessel hypertension. Consequently, despite differences in etiology, the pathophysiology of both chronic venous disease and diabetic microangiopathy appears to be driven by endothelial dysfunction and inflammation. Treatment strategies should take the co-existence of chronic venous disease and diabetic microangiopathy into account. Compression therapy is recommended in inflammatory conditions that have an edema component as seen in both chronic venous disease and diabetes mellitus. Lifestyle changes like weight loss and exercise, will improve metabolic state and lower inflammation and should be promoted in these patients. Additionally, both patient populations may benefit from venoactive drugs.
Collapse
Affiliation(s)
- Giacomo Gastaldi
- Division of Endocrinology Diabetology Nutrition and Patient Education, Geneva University Hospitals, Geneva, Switzerland
| | - Felizitas Pannier
- Private Clinic Phlebology and Dermatology, Bonn, Germany.,Department of Dermatology, University of Cologne, Cologne, Germany
| | - Karel Roztočil
- Department of Transplantational and Vascular Surgery, Institute for Clinical and Experimental Medicine, Prague, Czech Republic
| | - Marzia Lugli
- Unit of Vascular Surgery, Cardiovascular Department, Hesperia Hospital, Modena, Italy
| | - Armando Mansilha
- Faculty of Medicine, University of Porto, Porto, Portugal.,Department of Angiology and Vascular Surgery, Hospital de S. João, Porto, Portugal
| | - Hermann Haller
- Hannover Medical School, Department of Nephrology and Hypertension, Hannover, Germany
| | - Eberhard Rabe
- Department of Dermatology (Emeritus), University of Bonn, Bonn, Germany
| | - Marie Josee VAN Rijn
- Department of Vascular Surgery, Erasmus Medical Center, Rotterdam, the Netherlands -
| |
Collapse
|
10
|
Dallinga MG, Habani YI, Schimmel AWM, Dallinga-Thie GM, van Noorden CJF, Klaassen I, Schlingemann RO. The Role of Heparan Sulfate and Neuropilin 2 in VEGFA Signaling in Human Endothelial Tip Cells and Non-Tip Cells during Angiogenesis In Vitro. Cells 2021; 10:cells10040926. [PMID: 33923753 PMCID: PMC8073389 DOI: 10.3390/cells10040926] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Revised: 04/01/2021] [Accepted: 04/13/2021] [Indexed: 12/17/2022] Open
Abstract
During angiogenesis, vascular endothelial growth factor A (VEGFA) regulates endothelial cell (EC) survival, tip cell formation, and stalk cell proliferation via VEGF receptor 2 (VEGFR2). VEGFR2 can interact with VEGFR2 co-receptors such as heparan sulfate proteoglycans (HSPGs) and neuropilin 2 (NRP2), but the exact roles of these co-receptors, or of sulfatase 2 (SULF2), an enzyme that removes sulfate groups from HSPGs and inhibits HSPG-mediated uptake of very low density lipoprotein (VLDL), in angiogenesis and tip cell biology are unknown. In the present study, we investigated whether the modulation of binding of VEGFA to VEGFR2 by knockdown of SULF2 or NRP2 affects sprouting angiogenesis, tip cell formation, proliferation of non-tip cells, and EC survival, or uptake of VLDL. To this end, we employed VEGFA splice variant 121, which lacks an HSPG binding domain, and VEGFA splice variant 165, which does have this domain, in in vitro models of angiogenic tip cells and vascular sprouting. We conclude that VEGFA165 and VEGFA121 have similar inducing effects on tip cells and sprouting in vitro, and that the binding of VEGFA165 to HSPGs in the extracellular matrix does not seem to play a role, as knockdown of SULF2 did not alter these effects. Co-binding of NRP2 appears to regulate VEGFA–VEGFR2-induced sprout initiation, but not tip cell formation. Finally, as the addition of VLDL increased sprout formation but not tip cell formation, and as VLDL uptake was limited to non-tip cells, our findings suggest that VLDL plays a role in sprout formation by providing biomass for stalk cell proliferation.
Collapse
Affiliation(s)
- Marchien G. Dallinga
- Ocular Angiogenesis Group, Departments of Ophthalmology and Medical Biology, Amsterdam Cardiovascular Sciences, Cancer Center Amsterdam, Amsterdam UMC, University of Amsterdam, Meibergdreef 15, 1105 AZ Amsterdam, The Netherlands; (M.G.D.); (Y.I.H.); (C.J.F.v.N.); (R.O.S.)
| | - Yasmin I. Habani
- Ocular Angiogenesis Group, Departments of Ophthalmology and Medical Biology, Amsterdam Cardiovascular Sciences, Cancer Center Amsterdam, Amsterdam UMC, University of Amsterdam, Meibergdreef 15, 1105 AZ Amsterdam, The Netherlands; (M.G.D.); (Y.I.H.); (C.J.F.v.N.); (R.O.S.)
| | - Alinda W. M. Schimmel
- Department of Experimental Vascular Medicine, Amsterdam UMC, University of Amsterdam, Meibergdreef 15, 1105 AZ Amsterdam, The Netherlands; (A.W.M.S.); (G.M.D.-T.)
| | - Geesje M. Dallinga-Thie
- Department of Experimental Vascular Medicine, Amsterdam UMC, University of Amsterdam, Meibergdreef 15, 1105 AZ Amsterdam, The Netherlands; (A.W.M.S.); (G.M.D.-T.)
| | - Cornelis J. F. van Noorden
- Ocular Angiogenesis Group, Departments of Ophthalmology and Medical Biology, Amsterdam Cardiovascular Sciences, Cancer Center Amsterdam, Amsterdam UMC, University of Amsterdam, Meibergdreef 15, 1105 AZ Amsterdam, The Netherlands; (M.G.D.); (Y.I.H.); (C.J.F.v.N.); (R.O.S.)
- Department of Genetic Toxicology and Cancer Biology, National Institute of Biology, Večna pot 111, 1000 Ljubljana, Slovenia
| | - Ingeborg Klaassen
- Ocular Angiogenesis Group, Departments of Ophthalmology and Medical Biology, Amsterdam Cardiovascular Sciences, Cancer Center Amsterdam, Amsterdam UMC, University of Amsterdam, Meibergdreef 15, 1105 AZ Amsterdam, The Netherlands; (M.G.D.); (Y.I.H.); (C.J.F.v.N.); (R.O.S.)
- Correspondence:
| | - Reinier O. Schlingemann
- Ocular Angiogenesis Group, Departments of Ophthalmology and Medical Biology, Amsterdam Cardiovascular Sciences, Cancer Center Amsterdam, Amsterdam UMC, University of Amsterdam, Meibergdreef 15, 1105 AZ Amsterdam, The Netherlands; (M.G.D.); (Y.I.H.); (C.J.F.v.N.); (R.O.S.)
- Department of Ophthalmology, University of Lausanne, Jules Gonin Eye Hospital, Fondation Asile des Aveugles, Avenue de France 15, 1004 Lausanne, Switzerland
| |
Collapse
|
11
|
Zhang Q, Long J, Li N, Ma X, Zheng L. Circ_CLASP2 Regulates High Glucose-Induced Dysfunction of Human Endothelial Cells Through Targeting miR-140-5p/FBXW7 Axis. Front Pharmacol 2021; 12:594793. [PMID: 33776760 PMCID: PMC7990784 DOI: 10.3389/fphar.2021.594793] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Accepted: 01/18/2021] [Indexed: 01/10/2023] Open
Abstract
Hyperglycemia exposure results in the dysfunction of endothelial cells (ECs) and the development of diabetic complications. Circular RNAs (circRNAs) have been demonstrated to play critical roles in EC dysfunction. The current study aimed to explore the role and mechanism of circRNA CLIP–associating protein 2 (circ_CLASP2, hsa_circ_0064772) on HG-induced dysfunction in human umbilical vein endothelial cells (HUVECs). Quantitative real-time polymerase chain reaction (qRT-PCR) was used to assess the levels of circ_CLASP2, miR-140-5p and F-box, and WD repeat domain-containing 7 (FBXW7). The stability of circ_CLASP2 was identified by the actinomycin D and ribonuclease (RNase) R assays. Cell colony formation, proliferation, and apoptosis were measured by a standard colony formation assay, colorimetric 3-(4, 5-dimethylthiazol-2-yl)-2, 5-diphenyl-2H-tetrazolium bromide (MTT) assay, and flow cytometry, respectively. Western blot analysis was performed to determine the expression of related proteins. Targeted correlations among circ_CLASP2, miR-140-5p, and FBXW7 were confirmed by dual-luciferase reporter assay. High glucose (HG) exposure downregulated the expression of circ_CLASP2 in HUVECs. Circ_CLASP2 overexpression or miR-140-5p knockdown promoted proliferation and inhibited apoptosis of HUVECs under HG conditions. Circ_CLASP2 directly interacted with miR-140-5p via pairing to miR-140-5p. The regulation of circ_CLASP2 overexpression on HG-induced HUVEC dysfunction was mediated by miR-140-5p. Moreover, FBXW7 was a direct target of miR-140-5p, and miR-140-5p regulated HG-induced HUVEC dysfunction via FBXW7. Furthermore, circ_CLASP2 mediated FBXW7 expression through sponging miR-140-5p. Our current study suggested that the overexpression of circ_CLASP2 protected HUVEC from HG-induced dysfunction at least partly through the regulation of the miR-140-5p/FBXW7 axis, highlighting a novel therapeutic approach for the treatment of diabetic-associated vascular injury.
Collapse
Affiliation(s)
- Qin Zhang
- Department of Cardiovascular, Dongying People's Hospital, Dongying, China
| | - Jing Long
- Department of Critical Care Medicine, Dongying People's Hospital, Dongying, China
| | - Nannan Li
- Department of Cardiovascular, Dongying People's Hospital, Dongying, China
| | - Xuelian Ma
- Department of Clinical Laboratory, Dongying People's Hospital, Dongying, China
| | - Lisheng Zheng
- Department of Cardiovascular, Dongying People's Hospital, Dongying, China
| |
Collapse
|
12
|
Nensat C, Songjang W, Tohtong R, Suthiphongchai T, Phimsen S, Rattanasinganchan P, Metheenukul P, Kumphune S, Jiraviriyakul A. Porcine placenta extract improves high-glucose-induced angiogenesis impairment. BMC Complement Med Ther 2021; 21:66. [PMID: 33602182 PMCID: PMC7893890 DOI: 10.1186/s12906-021-03243-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Accepted: 02/09/2021] [Indexed: 11/29/2022] Open
Abstract
Background High glucose (HG)-induced reactive oxygen species (ROS) overproduction impairs angiogenesis that is one pivotal factor of wound healing process. Angiogenesis impairment induces delayed wound healing, whereby it eventually leads to amputation in cases of poorly controlled diabetes with diabetic ulceration. Porcine placenta extract (PPE) is a natural waste product that comprises plenty of bioactive agents including growth factors and antioxidants. It was reported as an effective compound that prevents ROS generation. The goal of this study was to investigate the in vitro effect of PPE on HG-induced ROS-mediated angiogenesis impairment. Methods Primary endothelial cells (HUVECs) and endothelial cell line (EA.hy926) were treated with HG in the presence of PPE. The endothelial cells (ECs) viability, intracellular ROS generation, migration, and angiogenesis were determined by MTT assay, DCFDA reagent, wound healing assay, and tube formation assay, respectively. Additionally, the molecular mechanism of PPE on HG-induced angiogenesis impairment was investigated by Western blot. The angiogenic growth factor secretion was also investigated by the sandwich ELISA technique. Results HG in the presence of PPE significantly decreased intracellular ROS overproduction compared to HG alone. HG in the presence of PPE significantly increased ECs viability, migration, and angiogenesis compared to HG alone by showing recovery of PI3K/Akt/ERK1/2 activation. HG in the presence of PPE also decreased ECs apoptosis compared to HG alone by decreasing p53/Bax/cleaved caspase 9/cleaved caspase 3 levels and increasing Bcl 2 level. Conclusion PPE attenuated HG-induced intracellular ROS overproduction that improved ECs viability, proliferation, migration, and angiogenesis by showing recovery of PI3K/Akt/ERK1/2 activation and inhibition of ECs apoptosis. This study suggests PPE ameliorated HG-induced ROS-mediated angiogenesis impairment, whereby it potentially provides an alternative treatment for diabetic wounds. Supplementary Information The online version contains supplementary material available at 10.1186/s12906-021-03243-z.
Collapse
Affiliation(s)
- Chatchai Nensat
- Integrative Biomedical Research Unit (IBRU), Faculty of Allied Health Sciences, Naresuan University, Phitsanulok, 65000, Thailand.,Graduate Program in Biomedical Sciences, Faculty of Allied Health Sciences, Naresuan University, Phitsanulok, 65000, Thailand
| | - Worawat Songjang
- Integrative Biomedical Research Unit (IBRU), Faculty of Allied Health Sciences, Naresuan University, Phitsanulok, 65000, Thailand.,Department of Medical Technology, Faculty of Allied Health Sciences, Naresuan University, Phitsanulok, 65000, Thailand
| | - Rutaiwan Tohtong
- Department of Biochemistry, Faculty of Science, Mahidol University, Bangkok, 10400, Thailand
| | | | - Suchada Phimsen
- Department of Biochemistry, Faculty of Medical Science, Naresuan University, Phitsanulok, 65000, Thailand
| | | | - Pornphimon Metheenukul
- Department of Veterinary Technology, Faculty of Veterinery Technology, Kasetsart University, Bangkok, 10900, Thailand
| | - Sarawut Kumphune
- Integrative Biomedical Research Unit (IBRU), Faculty of Allied Health Sciences, Naresuan University, Phitsanulok, 65000, Thailand.,Biomedical Engineering Institute (BMEI), Chiang Mai University, Chiang Mai, 50200, Thailand
| | - Arunya Jiraviriyakul
- Integrative Biomedical Research Unit (IBRU), Faculty of Allied Health Sciences, Naresuan University, Phitsanulok, 65000, Thailand. .,Department of Medical Technology, Faculty of Allied Health Sciences, Naresuan University, Phitsanulok, 65000, Thailand.
| |
Collapse
|
13
|
Morifuji T, Tanaka M, Nakanishi R, Hirabayashi T, Kondo H, Fujino H. Preventive effects of low-intensity endurance exercise for severe hyperglycemia-induced capillary regression in non-obese type 2 diabetes rat skeletal muscle. Physiol Rep 2021; 9:e14712. [PMID: 33463898 PMCID: PMC7814498 DOI: 10.14814/phy2.14712] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2020] [Revised: 12/10/2020] [Accepted: 12/12/2020] [Indexed: 12/12/2022] Open
Abstract
Although endurance exercise is effective for reducing diabetes‐related capillary regression, it is difficult to prescribe high‐intensity endurance exercise due to the potential worsening of complications in patients with severe hyperglycemia. Therefore, this study aimed to examine whether chronic low‐intensity exercise training may prevent severe hyperglycemia‐induced capillary regression of skeletal muscle in non‐obese type 2 diabetes. Non‐diabetic Sprague Dawley rats were assigned to a control (Con) group and an exercise (Ex) group. Likewise, spontaneously diabetic Torii rats were assigned to a diabetic sedentary (DM) group or a diabetic exercise (DMEx) group. Rats in the Ex and DMEx groups were placed on a motor‐driven treadmill running at low speed (15 m/min) for 60 min/day, 5 days/week, for 14 weeks. Serum glucose levels were significantly increased in the DM group, but not in the DMEx group. Although the capillary‐to‐fiber ratio in the plantaris muscle was significantly lower in the DM group compared to the control group, the ratio in the DMEx group was significantly higher compared to the DM group. Moreover, the succinate dehydrogenase activity and expression levels of vascular endothelial growth factor and peroxisome proliferator‐activated receptor γ coactivator‐1α (PGC‐1α) were reduced in the plantaris muscle of the DM group. However, those in the DMEx group were significantly higher than those in the DM group. These results indicate that low‐intensity chronic endurance exercise training has the potential to prevent the progression of capillary regression in the skeletal muscles of non‐obese type 2 diabetes patients with severe hyperglycemia.
Collapse
Affiliation(s)
- Takeshi Morifuji
- Department of Rehabilitation Science, Kobe University Graduate School of Health Sciences, Kobe, Japan.,Department of Physical Therapy, Josai International University, Tougane, Japan
| | - Minoru Tanaka
- Department of Rehabilitation Science, Kobe University Graduate School of Health Sciences, Kobe, Japan.,Department of Rehabilitation Science, Osaka Health Science University, Osaka, Japan
| | - Ryosuke Nakanishi
- Department of Rehabilitation Science, Kobe University Graduate School of Health Sciences, Kobe, Japan.,Department of Physical Therapy, Faculty of Rehabilitation, Kobe international University, Kobe, Japan
| | - Takumi Hirabayashi
- Department of Rehabilitation Science, Kobe University Graduate School of Health Sciences, Kobe, Japan
| | - Hiroyo Kondo
- Department of Food Science and Nutrition, Nagoya Women's University, Nagoya, Japan
| | - Hidemi Fujino
- Department of Rehabilitation Science, Kobe University Graduate School of Health Sciences, Kobe, Japan
| |
Collapse
|
14
|
Vascular Endothelial Growth Factor: A Translational View in Oral Non-Communicable Diseases. Biomolecules 2021; 11:biom11010085. [PMID: 33445558 PMCID: PMC7826734 DOI: 10.3390/biom11010085] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Revised: 01/08/2021] [Accepted: 01/08/2021] [Indexed: 02/06/2023] Open
Abstract
Vascular endothelial growth factors (VEGFs) are vital regulators of angiogenesis that are expressed in response to soluble mediators, such as cytokines and growth factors. Their physiologic functions include blood vessel formation, regulation of vascular permeability, stem cell and monocyte/macrophage recruitment and maintenance of bone homeostasis and repair. In addition, angiogenesis plays a pivotal role in chronic pathologic conditions, such as tumorigenesis, inflammatory immune diseases and bone loss. According to their prevalence, morbidity and mortality, inflammatory diseases affecting periodontal tissues and oral cancer are relevant non-communicable diseases. Whereas oral squamous cell carcinoma (OSCC) is considered one of the most common cancers worldwide, destructive inflammatory periodontal diseases, on the other hand, are amongst the most prevalent chronic inflammatory conditions affecting humans and also represent the main cause of tooth loss in adults. In the recent years, while knowledge regarding the role of VEGF signaling in common oral diseases is expanding, new potential translational applications emerge. In the present narrative review we aim to explore the role of VEGF signaling in oral cancer and destructive periodontal inflammatory diseases, with emphasis in its translational applications as potential biomarkers and therapeutic targets.
Collapse
|
15
|
Salvatore T, Pafundi PC, Galiero R, Rinaldi L, Caturano A, Vetrano E, Aprea C, Albanese G, Di Martino A, Ricozzi C, Imbriani S, Sasso FC. Can Metformin Exert as an Active Drug on Endothelial Dysfunction in Diabetic Subjects? Biomedicines 2020; 9:biomedicines9010003. [PMID: 33375185 PMCID: PMC7822116 DOI: 10.3390/biomedicines9010003] [Citation(s) in RCA: 71] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 12/18/2020] [Accepted: 12/21/2020] [Indexed: 12/12/2022] Open
Abstract
Cardiovascular mortality is a major cause of death among in type 2 diabetes (T2DM). Endothelial dysfunction (ED) is a well-known important risk factor for the development of diabetes cardiovascular complications. Therefore, the prevention of diabetic macroangiopathies by preserving endothelial function represents a major therapeutic concern for all National Health Systems. Several complex mechanisms support ED in diabetic patients, frequently cross-talking each other: uncoupling of eNOS with impaired endothelium-dependent vascular response, increased ROS production, mitochondrial dysfunction, activation of polyol pathway, generation of advanced glycation end-products (AGEs), activation of protein kinase C (PKC), endothelial inflammation, endothelial apoptosis and senescence, and dysregulation of microRNAs (miRNAs). Metformin is a milestone in T2DM treatment. To date, according to most recent EASD/ADA guidelines, it still represents the first-choice drug in these patients. Intriguingly, several extraglycemic effects of metformin have been recently observed, among which large preclinical and clinical evidence support metformin’s efficacy against ED in T2DM. Metformin seems effective thanks to its favorable action on all the aforementioned pathophysiological ED mechanisms. AMPK pharmacological activation plays a key role, with metformin inhibiting inflammation and improving ED. Therefore, aim of this review is to assess metformin’s beneficial effects on endothelial dysfunction in T2DM, which could preempt development of atherosclerosis.
Collapse
Affiliation(s)
- Teresa Salvatore
- Department of Precision Medicine, University of Campania Luigi Vanvitelli, Via De Crecchio 7, I-80138 Naples, Italy;
| | - Pia Clara Pafundi
- Department of Advanced Medical and Surgical Sciences, University of Campania Luigi Vanvitelli, Piazza Luigi Miraglia 2, I-80138 Naples, Italy; (P.C.P.); (R.G.); (L.R.); (A.C.); (E.V.); (C.A.); (G.A.); (A.D.M.); (C.R.); (S.I.)
| | - Raffaele Galiero
- Department of Advanced Medical and Surgical Sciences, University of Campania Luigi Vanvitelli, Piazza Luigi Miraglia 2, I-80138 Naples, Italy; (P.C.P.); (R.G.); (L.R.); (A.C.); (E.V.); (C.A.); (G.A.); (A.D.M.); (C.R.); (S.I.)
| | - Luca Rinaldi
- Department of Advanced Medical and Surgical Sciences, University of Campania Luigi Vanvitelli, Piazza Luigi Miraglia 2, I-80138 Naples, Italy; (P.C.P.); (R.G.); (L.R.); (A.C.); (E.V.); (C.A.); (G.A.); (A.D.M.); (C.R.); (S.I.)
| | - Alfredo Caturano
- Department of Advanced Medical and Surgical Sciences, University of Campania Luigi Vanvitelli, Piazza Luigi Miraglia 2, I-80138 Naples, Italy; (P.C.P.); (R.G.); (L.R.); (A.C.); (E.V.); (C.A.); (G.A.); (A.D.M.); (C.R.); (S.I.)
| | - Erica Vetrano
- Department of Advanced Medical and Surgical Sciences, University of Campania Luigi Vanvitelli, Piazza Luigi Miraglia 2, I-80138 Naples, Italy; (P.C.P.); (R.G.); (L.R.); (A.C.); (E.V.); (C.A.); (G.A.); (A.D.M.); (C.R.); (S.I.)
| | - Concetta Aprea
- Department of Advanced Medical and Surgical Sciences, University of Campania Luigi Vanvitelli, Piazza Luigi Miraglia 2, I-80138 Naples, Italy; (P.C.P.); (R.G.); (L.R.); (A.C.); (E.V.); (C.A.); (G.A.); (A.D.M.); (C.R.); (S.I.)
| | - Gaetana Albanese
- Department of Advanced Medical and Surgical Sciences, University of Campania Luigi Vanvitelli, Piazza Luigi Miraglia 2, I-80138 Naples, Italy; (P.C.P.); (R.G.); (L.R.); (A.C.); (E.V.); (C.A.); (G.A.); (A.D.M.); (C.R.); (S.I.)
| | - Anna Di Martino
- Department of Advanced Medical and Surgical Sciences, University of Campania Luigi Vanvitelli, Piazza Luigi Miraglia 2, I-80138 Naples, Italy; (P.C.P.); (R.G.); (L.R.); (A.C.); (E.V.); (C.A.); (G.A.); (A.D.M.); (C.R.); (S.I.)
| | - Carmen Ricozzi
- Department of Advanced Medical and Surgical Sciences, University of Campania Luigi Vanvitelli, Piazza Luigi Miraglia 2, I-80138 Naples, Italy; (P.C.P.); (R.G.); (L.R.); (A.C.); (E.V.); (C.A.); (G.A.); (A.D.M.); (C.R.); (S.I.)
| | - Simona Imbriani
- Department of Advanced Medical and Surgical Sciences, University of Campania Luigi Vanvitelli, Piazza Luigi Miraglia 2, I-80138 Naples, Italy; (P.C.P.); (R.G.); (L.R.); (A.C.); (E.V.); (C.A.); (G.A.); (A.D.M.); (C.R.); (S.I.)
| | - Ferdinando Carlo Sasso
- Department of Advanced Medical and Surgical Sciences, University of Campania Luigi Vanvitelli, Piazza Luigi Miraglia 2, I-80138 Naples, Italy; (P.C.P.); (R.G.); (L.R.); (A.C.); (E.V.); (C.A.); (G.A.); (A.D.M.); (C.R.); (S.I.)
- Correspondence: ; Tel.: +39-081-566-5010
| |
Collapse
|
16
|
Gong YP, Zhang YW, Su XQ, Gao HB. Inhibition of long noncoding RNA MALAT1 suppresses high glucose-induced apoptosis and inflammation in human umbilical vein endothelial cells by suppressing the NF-κB signaling pathway. Biochem Cell Biol 2020; 98:669-675. [PMID: 32502356 DOI: 10.1139/bcb-2019-0403] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
The study investigated the expression of long noncoding RNA (lncRNA) MALAT1 in high glucose (HG)-induced human vascular endothelial cells (HUVECs) and the role of MALAT1 in the apoptosis of HG-induced HUVECs. The HUVECs were cultured and induced with 25 mmol/L HG. After that, the HUVECs were transfected with MALAT1 siRNA. The expression levels of MALAT1 were detected with qPCR, whereas the expression levels of Bax, Bcl-2, cleaved-caspase-3, cleaved-caspase-9, p-65, and p-p65 were detected using Western blot. The roles of MALAT1 in cell activities, including apoptosis, were evaluated using the CCK-8 assay, TUNEL staining, and flow cytometry. The expression levels of inflammatory factors (TNF-α and IL-6) were measured using ELISA. The expression levels of MALAT1, TNF-α, and IL-6 in HUVECs were increased in the HG environment; however, when MALAT1 was silenced in the HUVECs, cell proliferation increased significantly, the expression levels of TNF-α, IL-6, Bax, cleaved-caspase-3, and cleaved-caspase-9 decreased, and the rate of apoptosis also decreased. Silencing MALAT1 inhibited the expression of p-p65 in HG-induced HUVECs. In conclusion, our study demonstrated that MALAT1 is upregulated in HG-induced HUVECs, and inhibition of MALAT1 inhibits HG-induced apoptosis and inflammation in HUVECs by suppression of the NF-κB signaling pathway.
Collapse
Affiliation(s)
- Yu-Ping Gong
- Department of Endocrinology, Pingxiang People's Hospital, Pingxiang 337000, P.R. China.,Department of Endocrinology, Pingxiang People's Hospital, Pingxiang 337000, P.R. China
| | - Ya-Wei Zhang
- Department of Endocrinology, Pingxiang People's Hospital, Pingxiang 337000, P.R. China.,Department of Endocrinology, Pingxiang People's Hospital, Pingxiang 337000, P.R. China
| | - Xiao-Qing Su
- Department of Endocrinology, Pingxiang People's Hospital, Pingxiang 337000, P.R. China.,Department of Endocrinology, Pingxiang People's Hospital, Pingxiang 337000, P.R. China
| | - Hai-Bo Gao
- Department of Endocrinology, Pingxiang People's Hospital, Pingxiang 337000, P.R. China.,Department of Endocrinology, Pingxiang People's Hospital, Pingxiang 337000, P.R. China
| |
Collapse
|
17
|
Fiori A, Hammes HP, Bieback K. Adipose-derived mesenchymal stromal cells reverse high glucose-induced reduction of angiogenesis in human retinal microvascular endothelial cells. Cytotherapy 2020; 22:261-275. [PMID: 32247542 DOI: 10.1016/j.jcyt.2020.02.005] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2019] [Revised: 02/12/2020] [Accepted: 02/22/2020] [Indexed: 12/28/2022]
Abstract
BACKGROUND AIMS Diabetic retinopathy (DR) is characterized by a progressive alteration of the retinal microvasculature, arising from microaneurysms to leaky vessels and finally abnormal neovascularization. The hyperglycemia-mediated loss of pericytes is a key event in vessel degeneration causing vascular destabilization. To overcome this, mesenchymal stromal cells (MSCs) have been tested as pericyte replacement in several animal models showing repair and regeneration of DR-damaged vasculature. METHODS We hypothesized that adipose-derived mesenchymal stromal cells (ASCs) resist high glucose-induced challenges and protect human retinal microvascular endothelial cells (HRMVECs) from glucose-mediated injury. ASCs and HRMVECs were cultured under normal-glucose (NG; 1 g/L) and high-glucose (HG; 4.5 g/L) conditions comparing their phenotype and angiogenic potential. RESULTS Whereas ASCs were generally unaffected by HG, HG caused a reduction of the angiogenic potential in HRMVEC. Indeed, HG-treated HRMVECs formed fewer vascular tube structures in a basement membrane angiogenesis assay. However, this was not observed in a direct ASC and HRMVEC coculture angiogenesis assay. Increased oxidative stress levels appeared to be linked to the HG-induced reduction of angiogenesis, which could be restored by ASC-conditioned medium and antioxidant treatment. CONCLUSIONS These findings suggest that ASC resist HG-stress whereas endothelial cell angiogenic capacity is reduced. Thus, ASC may be potentially therapeutically active in DR by restoring angiogenic deficits in retinal endothelial cells by the secretion of proangiogenic factors. However, these data also inquire for a thorough risk assessment about the timing of the ASC-based cell therapy, which can be considered advantageous at early stage of DR, but possibly detrimental at the late neo-angiogenic stage of DR.
Collapse
Affiliation(s)
- Agnese Fiori
- Institute of Transfusion Medicine and Immunology, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany, German Red Cross Blood Donor Service Baden-Württemberg-Hessen, Institute Mannheim, Germany
| | - Hans-Peter Hammes
- Endocrinology Department, 5th Medical Department, Medical Faculty Mannheim, Heidelberg University Mannheim, Baden-Württemberg, Germany
| | - Karen Bieback
- Institute of Transfusion Medicine and Immunology, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany, German Red Cross Blood Donor Service Baden-Württemberg-Hessen, Institute Mannheim, Germany; Flow Core Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Baden-Württemberg, Germany.
| |
Collapse
|
18
|
Liu J, Meng Z, Gan L, Guo R, Gao J, Liu C, Zhu D, Liu D, Zhang L, Zhang Z, Xie D, Jiao X, Lau WB, Lopez BL, Christopher TA, Ma X, Cao J, Wang Y. C1q/TNF-related protein 5 contributes to diabetic vascular endothelium dysfunction through promoting Nox-1 signaling. Redox Biol 2020; 34:101476. [PMID: 32122792 PMCID: PMC7327962 DOI: 10.1016/j.redox.2020.101476] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2019] [Revised: 02/19/2020] [Accepted: 02/21/2020] [Indexed: 12/19/2022] Open
Abstract
OBJECTIVE Dysregulated adipokine profiles contribute to the pathogenesis of diabetic cardiovascular complications. Endothelial cell (EC) dysfunction, a common pathological alteration in cardiovascular disorders, is exaggerated in diabetes. However, it is unclear whether and how dysregulated adipokines may contribute to diabetic EC dysfunction. METHODS AND RESULTS Serum C1q/TNF-Related Protein 5 (CTRP5) were determined in control/diabetes patients, and control/diabetic mice (high-fat diet, HFD). We observed for the first time that serum total CTRP5 was increased, high molecular weight (HMW) form was decreased, but the globular form (gCTRP5) was significantly increased in diabetic patients. These pathological alterations were reproduced in diabetic mice. To determine the pathological significance of increased gCTRP5 in diabetes, in vivo, ex vivo and in vitro experiments were performed. Diabetic atherosclerosis and EC dysfunction were significantly attenuated by the in vivo administration of CTRP5 neutralization antibody (CTRP5Ab). EC apoptosis was significantly increased in diabetic EC (isolated from HFD animal aorta) or high glucose high lipid (HGHL) cultured HUVECs. These pathological alterations were further potentiated by gCTRP5 and attenuated by CTRP5Ab. Pathway specific discovery-driven approach revealed that Nox1 expression was one of the signaling molecules commonly activated by HFD, HGHL, and gCTRP5. Treatment with CTRP5Ab reversed HFD-induced Nox1 upregulation. Finally, Nox1siRNA was used to determine the causative role of Nox1 in gCTRP5 induced EC apoptosis in diabetes. Results showed that gCTRP5 activated the mitochondrial apoptotic signal of EC in diabetes, which was blocked by the silencing Nox1 gene. CONCLUSION We demonstrated for the first time that gCTRP5 is a novel molecule contributing to diabetic vascular EC dysfunction through Nox1-mediated mitochondrial apoptosis, suggesting that interventions blocking gCTRP5 may protect diabetic EC function, ultimately attenuate diabetic cardiovascular complications.
Collapse
Affiliation(s)
- Jing Liu
- Department of Physiology, Shanxi Medical University, Shanxi, China; Department of Emergency Medicine, Thomas Jefferson University, Philadelphia, PA, USA
| | - Zhijun Meng
- Department of Physiology, Shanxi Medical University, Shanxi, China; Department of Emergency Medicine, Thomas Jefferson University, Philadelphia, PA, USA
| | - Lu Gan
- Department of Emergency Medicine, Thomas Jefferson University, Philadelphia, PA, USA
| | - Rui Guo
- Department of Physiology, Shanxi Medical University, Shanxi, China
| | - Jia Gao
- Department of Physiology, Shanxi Medical University, Shanxi, China
| | - Caihong Liu
- Department of Physiology, Shanxi Medical University, Shanxi, China
| | - Di Zhu
- Department of Emergency Medicine, Thomas Jefferson University, Philadelphia, PA, USA
| | - Demin Liu
- Department of Emergency Medicine, Thomas Jefferson University, Philadelphia, PA, USA
| | - Ling Zhang
- Department of Emergency Medicine, Thomas Jefferson University, Philadelphia, PA, USA
| | - Zhen Zhang
- Department of Emergency Medicine, Thomas Jefferson University, Philadelphia, PA, USA
| | - Dina Xie
- Department of Emergency Medicine, Thomas Jefferson University, Philadelphia, PA, USA
| | - Xiangying Jiao
- Department of Physiology, Shanxi Medical University, Shanxi, China
| | - Wayne Bond Lau
- Department of Emergency Medicine, Thomas Jefferson University, Philadelphia, PA, USA
| | - Bernard L Lopez
- Department of Emergency Medicine, Thomas Jefferson University, Philadelphia, PA, USA
| | | | - Xinliang Ma
- Department of Emergency Medicine, Thomas Jefferson University, Philadelphia, PA, USA
| | - Jimin Cao
- Department of Physiology, Shanxi Medical University, Shanxi, China.
| | - Yajing Wang
- Department of Emergency Medicine, Thomas Jefferson University, Philadelphia, PA, USA.
| |
Collapse
|
19
|
Joshi A, Gupta R, Singh B, Sharma D, Singh M. Effective inhibitory activity against MCF-7, A549 and HepG2 cancer cells by a phosphomolybdate based hybrid solid. Dalton Trans 2020; 49:7069-7077. [DOI: 10.1039/d0dt01042a] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
A novel P2Mo5 cluster based hybrid solid [{4,4′-H2bpy}{4,4′-Hbpy}2{H2P2Mo5O23}]·5H2O with effective anti-proliferation activity against MCF-7, HepG2 and A549 cancer cells comparable with a routinely used chemotherapeutic agent, methotrexate (MTX).
Collapse
Affiliation(s)
- Arti Joshi
- Institute of Nano Science and Technology
- Mohali-160062
- India
| | - Ruby Gupta
- Institute of Nano Science and Technology
- Mohali-160062
- India
| | - Bharti Singh
- Department of Chemistry
- Indian Institute of Technology Delhi
- New Delhi-110016
- India
| | - Deepika Sharma
- Institute of Nano Science and Technology
- Mohali-160062
- India
| | - Monika Singh
- Institute of Nano Science and Technology
- Mohali-160062
- India
| |
Collapse
|
20
|
Lechner J, Hombrebueno JR, Pedrini E, Chen M, Xu H. Sustained intraocular vascular endothelial growth factor neutralisation does not affect retinal and choroidal vasculature in Ins2 Akita diabetic mice. Diab Vasc Dis Res 2019; 16:440-449. [PMID: 31023085 DOI: 10.1177/1479164119843092] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
The purpose of this study was to understand the influence of sustained intravitreal vascular endothelial growth factor neutralisation on the retinal and choroidal vasculature in diabetic eyes. Ins2Akita diabetic mice received five intravitreal injections of anti-mouse vascular endothelial growth factor antibody or goat immunoglobulin G (0.2 µg/µL/eye) over a 4-month period. Retinal and choroidal vascular changes were analysed by confocal microscopy of tissue flat-mounts. Retinal gene expression of vascular endothelial growth factor family members (vascular endothelial growth factors A, B, C and D), vascular endothelial growth factor receptors (sVEGFR-1 and VEGFR-2) and tight junctions (claudin 1, 2, 5; occludin and zonula occludens-1) were analysed by quantitative reverse transcription polymerase chain reaction. Vascular endothelial growth factor A and claudin 5 were significantly increased in diabetic retinae. Gene expression was unaffected by anti-vascular endothelial growth factor treatment. The number of acellular vessels was increased in diabetic retinae and reduced following anti-vascular endothelial growth factor treatment. Retinal and choroidal vascular density and area were unaffected by sustained vascular endothelial growth factor neutralisation. Our results suggest that five consecutive intravitreal anti-vascular endothelial growth factor injections do not cause significant vascular changes in the retina and choroid in diabetic and non-diabetic mice.
Collapse
Affiliation(s)
- Judith Lechner
- Centre for Experimental Medicine, Queen's University Belfast, Belfast, UK
| | - Jose R Hombrebueno
- Centre for Experimental Medicine, Queen's University Belfast, Belfast, UK
| | - Edoardo Pedrini
- Centre for Experimental Medicine, Queen's University Belfast, Belfast, UK
| | - Mei Chen
- Centre for Experimental Medicine, Queen's University Belfast, Belfast, UK
| | - Heping Xu
- Centre for Experimental Medicine, Queen's University Belfast, Belfast, UK
| |
Collapse
|
21
|
Dabrowski FA, Lipa M, Bartoszewicz Z, Wielgos M, Bomba-Opon DA. Maternal and neonatal serum expression of the vascular growth factors in hyperglycemia in pregnancy. J Matern Fetal Neonatal Med 2019; 34:1673-1678. [PMID: 31307255 DOI: 10.1080/14767058.2019.1639666] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
INTRODUCTION Hyperglycemia in pregnancy (HIP) occurs in up to 8-17% of pregnancies. Unfavorable impact of the pregnancy induced hyperglycemia on both maternal and fetal tissues is associated with adverse pregnancy outcomes. Vascular growth factors, especially in the early phase of gestation, are considered as one of the most significant molecules that regulate pregnancy course and their serum expression may be altered in patients affected with HIP. MATERIAL AND METHODS Fifty-five consecutive pregnant patients who underwent elective cesarean section were incorporated into this study. During the surgery, maternal and cord blood samples were collected. Serum expression levels of vascular growth factors: PlGF, VEGF, THBS-2 and Ang-2 were compared among non-HIP and pregnancies affected by gestational diabetes. Subsequently, laboratory results were correlated with obstetric outcomes. RESULTS There were no statistical differences in maternal characteristics, neonatal outcomes and maternal or neonatal serum levels between study and control groups. However, our results revealed significant differences between fetal and maternal levels of VEGF (p = .028 and .0001), THBS-2 (p = .013 and .0014) and Ang-2 (p = .035 and .048) for HIP and non-HIP group, respectively. CONCLUSIONS Similar serum expressions of vascular growth factors in and non-HIP and HIP pregnancies point that normal glycemia due to thorough prenatal surveillance may result in normal angio- and vasculogenesis associated with good pregnancy outcomes.
Collapse
Affiliation(s)
- Filip A Dabrowski
- 1st Department of Obstetrics and Gynecology, 1st Faculty of Medicine, Medical University of Warsaw, Warsaw, Poland
| | - Michal Lipa
- 1st Department of Obstetrics and Gynecology, 1st Faculty of Medicine, Medical University of Warsaw, Warsaw, Poland
| | - Zbigniew Bartoszewicz
- Department of Endocrinology and Internal Medicine, 1st Faculty of Medicine, Medical University of Warsaw, Warsaw, Poland
| | - Miroslaw Wielgos
- 1st Department of Obstetrics and Gynecology, 1st Faculty of Medicine, Medical University of Warsaw, Warsaw, Poland
| | - Dorota A Bomba-Opon
- 1st Department of Obstetrics and Gynecology, 1st Faculty of Medicine, Medical University of Warsaw, Warsaw, Poland
| |
Collapse
|
22
|
Caprnda M, Kubatka P, Saxena S, Valaskova J, Stefanickova J, Kobyliak N, Zulli A, Kruzliak P. The Impact of Hyperglycemia on VEGF Secretion in Retinal Endothelial Cells. Folia Med (Plovdiv) 2019; 59:183-189. [PMID: 28704181 DOI: 10.1515/folmed-2017-0029] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2016] [Accepted: 12/21/2016] [Indexed: 11/15/2022] Open
Abstract
BACKGROUND Diabetic retinopathy is a serious sight-threatening complication which is manifested by excessive angiogenesis in diabetic patients. AIM We hypothesize that cultured Rhesus monkey retinal endothelial cells (RhRECs) respond to high glucose with a change in cell proliferation and vascular endothelial growth factor (VEGF) secretion. MATERIALS AND METHODS In our study, 20 000 cells per well were treated without glucose or with 5.5 mM, 18.5 mM and 30 mM glucose for 24 hours. Viable cells were counted using trypan blue dye exclusion method. VEGF concentrations were measured in cell media by ELISA method. RESULTS The number of viable cells incubated with 5.5 mM glucose increased significantly by 53.7% after 24 hours. In comparison, the number of viable cells decreased by 2.8% at 18.5 mM of glucose and by 20.4% at 30 mM of glucose after 24 hours of incubation. In contrast to this effect of glucose on the number of viable cells, a significant increase in VEGF levels (pg/mL) in the cell media with a glucose concentration of 0 mM compared to 5.5 mM of glucose was found. VEGF secretion in cell medium with 18.5 and 30 mM of glucose increased non-significantly in comparison with euglycemic levels. CONCLUSION Our results show that viability of retinal endothelial cells and VEGF release are highly responsive to changes in glucose concentration. Such glucose-induced changes in retinal endothelial cells may negatively impact the integrity of the microvasculature in the diabetic retina leading to angiogenesis and microaneursym.
Collapse
Affiliation(s)
- Martin Caprnda
- First Department of Internal Medicine, Faculty of Medicine, Comenius University, Bratislava, Slovakia
| | - Peter Kubatka
- Department of Medical Biology, Jessenius Faculty of Medicine, Comenius University in Bratislava, Martin, Slovak Republic
| | - Sandeep Saxena
- Retina service, Department of Ophthalmology, King George’s Medical University, Lucknow, India
| | - Jela Valaskova
- Department of Ophthalmology, Faculty of Medicine, Comenius University and University Hospital, Bratislava, Slovakia
| | - Jana Stefanickova
- Department of Ophthalmology, Faculty of Medicine, Comenius University and University Hospital, Bratislava, Slovakia
| | - Nazarii Kobyliak
- Endocrinology Department, Bogomolets National Medical University, Kyiv, Ukraine
| | - Anthony Zulli
- Centre for Chronic Disease (CCD), College of Health & Biomedicine, Victoria University, Melbourne, Victoria, Australia
| | - Peter Kruzliak
- Department of Chemical Drugs, Faculty of Pharmacy, University of Veterinary and Pharmaceutical Sciences, Brno, Czech
Republic
| |
Collapse
|
23
|
Xu J, Liu M, Yu M, Shen J, Zhou J, Hu J, Zhou Y, Zhang W. RasGRP1 is a target for VEGF to induce angiogenesis and involved in the endothelial‐protective effects of metformin under high glucose in HUVECs. IUBMB Life 2019; 71:1391-1400. [PMID: 31120617 DOI: 10.1002/iub.2072] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2019] [Accepted: 04/30/2019] [Indexed: 12/14/2022]
Affiliation(s)
- Jing Xu
- Department of Clinical PharmacologyXiangya Hospital, Central South University Changsha People's Republic of China
- Institute of Clinical Pharmacology, Central South UniversityHunan Key Laboratory of Pharmacogenetics Changsha People's Republic of China
| | - Miao Liu
- Department of Clinical PharmacologyXiangya Hospital, Central South University Changsha People's Republic of China
- Institute of Clinical Pharmacology, Central South UniversityHunan Key Laboratory of Pharmacogenetics Changsha People's Republic of China
| | - Muqiao Yu
- Center of StomatologyXiangya Hospital, Central South University Changsha Hunan People's Republic of China
| | - Jiayi Shen
- Department of Clinical PharmacologyXiangya Hospital, Central South University Changsha People's Republic of China
- Institute of Clinical Pharmacology, Central South UniversityHunan Key Laboratory of Pharmacogenetics Changsha People's Republic of China
| | - Jiecan Zhou
- Department of Clinical PharmacologyXiangya Hospital, Central South University Changsha People's Republic of China
- Institute of Clinical Pharmacology, Central South UniversityHunan Key Laboratory of Pharmacogenetics Changsha People's Republic of China
| | - Jinglei Hu
- Department of Clinical PharmacologyXiangya Hospital, Central South University Changsha People's Republic of China
- Institute of Clinical Pharmacology, Central South UniversityHunan Key Laboratory of Pharmacogenetics Changsha People's Republic of China
| | - Yong Zhou
- Department of OrthopaediesThe Third Xiangya Hospital, Central South University Changsha Hunan People's Republic of China
| | - Wei Zhang
- Department of Clinical PharmacologyXiangya Hospital, Central South University Changsha People's Republic of China
- Institute of Clinical Pharmacology, Central South UniversityHunan Key Laboratory of Pharmacogenetics Changsha People's Republic of China
- Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education Changsha People's Republic of China
- National Clinical Research Center for Geriatric Disorders Changsha Hunan People's Republic of China
| |
Collapse
|
24
|
Nafisa A, Gray SG, Cao Y, Wang T, Xu S, Wattoo FH, Barras M, Cohen N, Kamato D, Little PJ. Endothelial function and dysfunction: Impact of metformin. Pharmacol Ther 2018; 192:150-162. [PMID: 30056057 DOI: 10.1016/j.pharmthera.2018.07.007] [Citation(s) in RCA: 91] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Cardiovascular and metabolic diseases remain the leading cause of morbidity and mortality worldwide. Endothelial dysfunction is a key player in the initiation and progression of cardiovascular and metabolic diseases. Current evidence suggests that the anti-diabetic drug metformin improves insulin resistance and protects against endothelial dysfunction in the vasculature. Hereby, we provide a timely review on the protective effects and molecular mechanisms of metformin in preventing endothelial dysfunction and cardiovascular and metabolic diseases.
Collapse
Affiliation(s)
- Asma Nafisa
- School of Pharmacy, University of Queensland, Pharmacy Australia Centre of Excellence, Woolloongabba, QLD, Australia.
| | - Susan G Gray
- School of Pharmacy, University of Queensland, Pharmacy Australia Centre of Excellence, Woolloongabba, QLD, Australia.
| | - Yingnan Cao
- Xinhua College of Sun Yat-sen University, Tianhe District, Guangzhou, China
| | - Tinghuai Wang
- Xinhua College of Sun Yat-sen University, Tianhe District, Guangzhou, China.
| | - Suowen Xu
- Aab Cardiovascular Research Institute, Department of Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA.
| | - Feroza H Wattoo
- Department of Biochemistry, PMAS Arid Agriculture University, Shamasabad, Muree Road, Rawalpindi 4600, Pakistan..
| | - Michael Barras
- Dept. of Pharmacy, Princess Alexandra Hospital, 199 Ipswich Rd, Woolloongabba, QLD 4102, Australia.
| | - Neale Cohen
- Baker Heart and Diabetes Institute, Melbourne, 3004, Victoria, Australia.
| | - Danielle Kamato
- School of Pharmacy, University of Queensland, Pharmacy Australia Centre of Excellence, Woolloongabba, QLD, Australia; Xinhua College of Sun Yat-sen University, Tianhe District, Guangzhou, China.
| | - Peter J Little
- School of Pharmacy, University of Queensland, Pharmacy Australia Centre of Excellence, Woolloongabba, QLD, Australia; Xinhua College of Sun Yat-sen University, Tianhe District, Guangzhou, China.
| |
Collapse
|
25
|
Alshamsan A. STAT3-siRNA induced B16.F10 melanoma cell death: more association with VEGF downregulation than p-STAT3 knockdown. Saudi Pharm J 2018; 26:1083-1088. [PMID: 30532628 PMCID: PMC6260487 DOI: 10.1016/j.jsps.2018.05.018] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2018] [Accepted: 05/29/2018] [Indexed: 12/14/2022] Open
Abstract
STAT3 knockdown by small interfering RNA (siRNA) has been described to inhibit carcinogenic growth in various types of tumors. Earlier we have reported delivery of siRNA by oleic acid- and stearic acid-modified-polyethylenimine and enhancement of silencing of STAT3 by small interfering RNA (siRNA) in B16.F10 melanoma cell lines and consequent tumor suppression. Present investigation mainly focused on the downstream events involved in B16.F10 melanoma cell death and consequent tumor suppression following knockdown of p-STAT3 by siRNA. Lipid-substituted polyethylenimine (PEI)-p-STAT3-siRNA were prepared and characterized by measuring its N/P ratio, zeta potential, size, association and dissociation with siRNA. B16.F10 melanoma cells were treated with six different concentrations of PEI-p-STAT3-siRNA (200, 100, 50, 25, 12.5 and 6.25 nM). Downregulation of p-STAT3 and VEGF were studied using western blot and ELISA in association with the melanoma cell death. PEI-p-STAT3-siRNA hydrodynamic diameter ranged from 110 to 270 nm. PEI assisted p-STAT3-siRNA delivery exhibited increased uptake by B16.F10, when analyzed by fluorescent and confocal microscopy along with flowcytometry. It induced concentration-dependent knockdown of the p-STAT3 that also downregulated VEGF expression in similar fashion and induced B16.F10 cell death. Higher concentrations of p-STAT3-siRNA appear to significantly downregulate the VEGF expression via p-STAT3 knockdown. Decreasing survival of B16.F10 cells with the increasing concentration of p-STAT3-siRNA significantly correlated with VEGF downregulation, not with p-STAT3 expression. Data suggest that VEGF downregulation following knockdown of p-STAT3 may be a key event in survival reduction in B16.F10 melanoma cells and.
Collapse
Affiliation(s)
- Aws Alshamsan
- Nanomedicine Research Unit, Department of Pharmaceutics, College of Pharmacy, King Saud University, Saudi Arabia.,King Abdullah Institute for Nanotechnology, King Saud University, Saudi Arabia
| |
Collapse
|
26
|
Qing Z, Xiao-Hui W, Xi-Mei W, Chao-Chun Z. Vitamin C deficiency aggravates tumor necrosis factor α-induced insulin resistance. Eur J Pharmacol 2018; 829:1-11. [PMID: 29625084 DOI: 10.1016/j.ejphar.2018.03.044] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2018] [Revised: 03/26/2018] [Accepted: 03/29/2018] [Indexed: 02/06/2023]
Abstract
Chronic low-grade inflammation plays a major role in the development of insulin resistance. The potential role and underlying mechanism of vitamin C, an antioxidant and anti-inflammatory agent, was investigated in tumor necrosis factor-α (TNF-α)-induced insulin resistance. Gulonolactone oxidase knockout (Gulo-/-) mice genetically unable to synthesize vitamin C were used to induce insulin resistance by continuously pumping small doses of TNF-α for seven days, and human liver hepatocellular carcinoma cells (HepG2 cells) were used to induce insulin resistance by treatment with TNF-α. Vitamin C deficiency aggravated TNF-α-induced insulin resistance in Gulo-/- mice, resulting in worse glucose tolerance test (GTT) results, higher fasting plasma insulin level, and the inactivation of the protein kinase B (AKT)/glycogen synthase kinase-3β (GSK3β) pathway in the liver. Vitamin C deficiency also worsened liver lipid accumulation and inflammation in TNF-α-treated Gulo-/- mice. In HepG2 cells, vitamin C reversed the TNF-α-induced reduction of glucose uptake and glycogen synthesis, which were mediated by increasing GLUT2 levels and the activation of the insulin receptor substrate (IRS-1)/AKT/GSK3β pathway. Furthermore, vitamin C inhibited the TNF-α-induced activation of not only the mitogen-activated protein kinase (MAPKs), but also nuclear factor-kappa B (NF-κB) signaling. Taken together, vitamin C is essential for preventing and improving insulin resistance, and the supplementing with vitamin C may be an effective therapeutic intervention for metabolic disorders.
Collapse
Affiliation(s)
- Zhou Qing
- Department of Endocrinology of the affiliated Children's Hospital, Zhejiang University School of Medicine, China
| | - Wu Xiao-Hui
- Department of Endocrinology of the affiliated Children's Hospital, Zhejiang University School of Medicine, China
| | - Wu Xi-Mei
- Department of Pharmacology, Zhejiang University School of Medicine, China
| | - Zou Chao-Chun
- Department of Endocrinology of the affiliated Children's Hospital, Zhejiang University School of Medicine, China.
| |
Collapse
|
27
|
Watson EC, Grant ZL, Coultas L. Endothelial cell apoptosis in angiogenesis and vessel regression. Cell Mol Life Sci 2017; 74:4387-4403. [PMID: 28646366 PMCID: PMC11107683 DOI: 10.1007/s00018-017-2577-y] [Citation(s) in RCA: 99] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2017] [Revised: 06/14/2017] [Accepted: 06/16/2017] [Indexed: 12/11/2022]
Abstract
Blood vessel regression is an essential process for ensuring blood vessel networks function at optimal efficiency and for matching blood supply to the metabolic needs of tissues as they change over time. Angiogenesis is the major mechanism by which new blood vessels are produced, but the vessel growth associated with angiogenesis must be complemented by remodeling and maturation events including the removal of redundant vessel segments and cells to fashion the newly forming vasculature into an efficient, hierarchical network. This review will summarize recent findings on the role that endothelial cell apoptosis plays in vascular remodeling during angiogenesis and in vessel regression more generally.
Collapse
Affiliation(s)
- Emma C Watson
- Development and Cancer Division, The Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade, Parkville, VIC, 3052, Australia
- Department of Medical Biology, University of Melbourne, 1G Royal Parade, Parkville, VIC, 3052, Australia
- Department of Tissue Morphogenesis, Max Planck Institute for Molecular Biomedicine, 48149, Münster, Germany
- Faculty of Medicine, University of Münster, 48149, Münster, Germany
| | - Zoe L Grant
- Development and Cancer Division, The Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade, Parkville, VIC, 3052, Australia
- Department of Medical Biology, University of Melbourne, 1G Royal Parade, Parkville, VIC, 3052, Australia
| | - Leigh Coultas
- Development and Cancer Division, The Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade, Parkville, VIC, 3052, Australia.
- Department of Medical Biology, University of Melbourne, 1G Royal Parade, Parkville, VIC, 3052, Australia.
| |
Collapse
|
28
|
Osuka K, Watanabe Y, Usuda N, Aoyama M, Iwami K, Takeuchi M, Watabe T, Takayasu M. Expression of Caspase Signaling Components in the Outer Membranes of Chronic Subdural Hematomas. J Neurotrauma 2017; 34:3192-3197. [DOI: 10.1089/neu.2017.5051] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Affiliation(s)
- Koji Osuka
- Department of Neurological Surgery, Aichi Medical University, Nagakute, Aichi, Japan
| | - Yasuo Watanabe
- High Technology Research Center, Pharmacology, Showa Pharmaceutical University, Machida, Tokyo, Japan
| | - Nobuteru Usuda
- Department of Anatomy II, Fujita Health University School of Medicine, Kutsukake, Toyoake, Aichi, Japan
| | - Masahiro Aoyama
- Department of Neurological Surgery, Aichi Medical University, Nagakute, Aichi, Japan
| | - Kenichiro Iwami
- Department of Neurological Surgery, Aichi Medical University, Nagakute, Aichi, Japan
| | - Mikinobu Takeuchi
- Department of Neurological Surgery, Aichi Medical University, Nagakute, Aichi, Japan
| | - Takeya Watabe
- Department of Neurological Surgery, Aichi Medical University, Nagakute, Aichi, Japan
| | - Masakazu Takayasu
- Department of Neurological Surgery, Aichi Medical University, Nagakute, Aichi, Japan
| |
Collapse
|
29
|
Martín-Saavedra F, Crespo L, Escudero-Duch C, Saldaña L, Gómez-Barrena E, Vilaboa N. Substrate Microarchitecture Shapes the Paracrine Crosstalk of Stem Cells with Endothelial Cells and Osteoblasts. Sci Rep 2017; 7:15182. [PMID: 29123118 PMCID: PMC5680323 DOI: 10.1038/s41598-017-15036-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2017] [Accepted: 10/19/2017] [Indexed: 01/08/2023] Open
Abstract
We examined the hypothesis that substrate microarchitecture regulates the crosstalk between human mesenchymal stem cells (hMSC) and cell types involved in bone regeneration. Compared with polyester flat substrates having uniformly distributed homogenous pores (2D), three-dimensional polystyrene substrates with randomly oriented and interconnected pores of heterogeneous size (3D) stimulated the stromal secretion of IGF-1 while lessened the production of VEGFR-1, MCP-1 and IL-6. The medium conditioned by hMSC cultured in 3D substrates stimulated tube formation by human endothelial cells (hEC) to a higher extent than medium from 2D cultures. 3D co-cultures of hMSC and hEC contained higher secreted levels of IGF-1, EGF and FGF-2 than 2D co-cultures, resulting in increased hEC proliferation and migration. Substrate microarchitecture influenced the secretion of factors related to bone remodeling as the ratio RANKL to OPG, and the levels of M-CSF and IL-6 were higher in 3D co-cultures of hMSC and human osteoblasts (hOB) than in 2D co-cultures. Cytokine microenvironment in 3D co-cultures stimulated osteoblast matrix reorganization while demoted the late steps of osteoblastic maturation. Altogether, data in this study may unveil a new role of scaffold microarchitecture during bone regeneration, as modulator of the paracrine relationships that hMSC establish with hEC and hOB.
Collapse
Affiliation(s)
- Francisco Martín-Saavedra
- CIBER de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Madrid, Spain.,Hospital Universitario La Paz-IdiPAZ, Paseo de la Castellana 261, 28046, Madrid, Spain
| | - Lara Crespo
- Hospital Universitario La Paz-IdiPAZ, Paseo de la Castellana 261, 28046, Madrid, Spain
| | - Clara Escudero-Duch
- CIBER de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Madrid, Spain.,Hospital Universitario La Paz-IdiPAZ, Paseo de la Castellana 261, 28046, Madrid, Spain
| | - Laura Saldaña
- CIBER de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Madrid, Spain.,Hospital Universitario La Paz-IdiPAZ, Paseo de la Castellana 261, 28046, Madrid, Spain
| | - Enrique Gómez-Barrena
- Hospital Universitario La Paz-IdiPAZ, Paseo de la Castellana 261, 28046, Madrid, Spain.,Departamento de Cirugía, Universidad Autónoma de Madrid, Calle del Arzobispo Morcillo 4, 28029, Madrid, Spain
| | - Nuria Vilaboa
- CIBER de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Madrid, Spain. .,Hospital Universitario La Paz-IdiPAZ, Paseo de la Castellana 261, 28046, Madrid, Spain.
| |
Collapse
|
30
|
Hassanpour M, Rezabakhsh A, Rahbarghazi R, Nourazarian A, Nouri M, Avci ÇB, Ghaderi S, Alidadyani N, Bagca BG, Bagheri HS. Functional convergence of Akt protein with VEGFR-1 in human endothelial progenitor cells exposed to sera from patient with type 2 diabetes mellitus. Microvasc Res 2017; 114:101-113. [PMID: 28732797 DOI: 10.1016/j.mvr.2017.07.002] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2017] [Revised: 06/27/2017] [Accepted: 07/17/2017] [Indexed: 11/18/2022]
Abstract
Diabetes mellitus type 2 predisposes patients to various microvascular complications. In the current experiment, the potent role of diabetes mellitus was investigated on the content of VEGFR-1, -2, Tie-1 and -2, and Akt in human endothelial progenitor cells. The gene expression profile of mTOR and Hedgehog signaling pathways were measured by PCR array. The possible crosstalk between RTKs, mTOR and Hedgehog signaling was also studied by bioinformatic analysis. Endothelial progenitor cells were incubated with serum from normal and diabetic for 7days. Compared to non-treated cells, diabetic serum-induced cell apoptosis (~2-fold) and prohibited cell migration toward bFGF (p<0.001). ELISA analysis showed that diabetes exposed cells had increased abundance of Tie-1, -2 and VEGFR-2 and reduced amount of VEGFR-1 (p<0.0001) in diabetic cells. Western blotting showed a marked reduction in the protein level of Akt after cells exposure to serum from diabetic subjects (p<0.0001). PCR array revealed a significant stimulation of both mTOR and Hedgehog signaling pathways in diabetic cells (p<0.05). According to data from bioinformatic datasets, we showed VEGFR-1, -2 and Tie-2, but not Tie-1, are master regulators of angiogenesis. There is a crosstalk between RTKs and mTOR signaling by involving P62, GABARAPL1, and HTT genes. It seems that physical interaction and co-expression of Akt decreased the level of VEGFR-1 in diabetic cells. Regarding data from the present experiment, diabetic serum contributed to uncontrolled induction of both mTOR and Hedgehog signaling in endothelial progenitor cells. Diabetes mellitus induces mTOR pathway by involving receptor tyrosine kinases while Hedgehog stimulation is independent of these receptors.
Collapse
Affiliation(s)
- Mehdi Hassanpour
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Applied Cell Sciences, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Aysa Rezabakhsh
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Pharmacology and Toxicology, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Reza Rahbarghazi
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Applied Cell Sciences, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Alireza Nourazarian
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Biochemistry and Clinical Laboratories, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Mohammad Nouri
- Department of Biochemistry and Clinical Laboratories, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Çığır Biray Avci
- Faculty of Medicine, Department of Medical Biology, Ege University, Izmir, Turkey.
| | - Shahrooz Ghaderi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Molecular Medicine, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Neda Alidadyani
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Bakiye Goker Bagca
- Faculty of Medicine, Department of Medical Biology, Ege University, Izmir, Turkey
| | | |
Collapse
|
31
|
Wang D, Xin Y, Tian Y, Li W, Sun D, Yang Y. Pseudolaric acid B inhibits gastric cancer cell metastasis in vitro and in haematogenous dissemination model through PI3K/AKT, ERK1/2 and mitochondria-mediated apoptosis pathways. Exp Cell Res 2017; 352:34-44. [PMID: 28132880 DOI: 10.1016/j.yexcr.2017.01.012] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2016] [Revised: 01/15/2017] [Accepted: 01/25/2017] [Indexed: 12/30/2022]
Abstract
Pseudolaric acid B (PAB) is the major bioactive constituent in the root bark of Pseudolarix kaempferi and has been reported to have cytotoxicity against tumor cells. Our in vivo experiments showed that PAB could inhibit gastric cancer cell lung metastasis in a nude mouse haematogenous dissemination model. To evaluate the anti-metastasis mechanism of PAB in gastric cancer cells, cytological experiments were performed. The results showed that PAB could inhibit the adhesion ability to matrigel, migration, invasion and colony formation ability of BGC-823 and MKN-45 cells. Western blot further confirmed that the inhibitory effects of PAB on anti-metastasis may involve regulating the expression of the metastasis-related proteins MMP-9, HIF-1α, VEGF, VEGFR2, E-Cadherin and Ezrin. We obtained further proof that PAB which could be used as a multi-targeted agent to inhibit the PI3K/AKT, ERK1/2 and mitochondria-mediated apoptosis pathways and consequently suppress tumor growth and metastasis. Our experiments suggest that PAB-induced effects may have novel therapeutic applications for the treatment of gastric cancer.
Collapse
Affiliation(s)
- Dan Wang
- Department of Gastrointestinal Tumor Pathology of Cancer Institute and General Surgery Institute, The First Hospital of China Medical University, Shenyang, China; Colledge of Pharmacy, Liaoning University, Shenyang, China.
| | - Yan Xin
- Department of Gastrointestinal Tumor Pathology of Cancer Institute and General Surgery Institute, The First Hospital of China Medical University, Shenyang, China
| | - Yanqiu Tian
- Colledge of Life Science, Liaoning University, Shenyang, China
| | - Wenhui Li
- Department of Gastrointestinal Tumor Pathology of Cancer Institute and General Surgery Institute, The First Hospital of China Medical University, Shenyang, China
| | - Dan Sun
- Department of Gastrointestinal Tumor Pathology of Cancer Institute and General Surgery Institute, The First Hospital of China Medical University, Shenyang, China
| | - Yi Yang
- Laboratory Animal Center, China Medical University, Shenyang, China
| |
Collapse
|
32
|
Howell DW, Duran CL, Tsai SP, Bondos SE, Bayless KJ. Functionalization of Ultrabithorax Materials with Vascular Endothelial Growth Factor Enhances Angiogenic Activity. Biomacromolecules 2016; 17:3558-3569. [DOI: 10.1021/acs.biomac.6b01068] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Affiliation(s)
- David W. Howell
- Department of Molecular and Cellular Medicine, Texas A&M University Health Science Center, College Station, Texas 77843, United States
| | - Camille L. Duran
- Department of Molecular and Cellular Medicine, Texas A&M University Health Science Center, College Station, Texas 77843, United States
| | - Shang-Pu Tsai
- Department of Molecular and Cellular Medicine, Texas A&M University Health Science Center, College Station, Texas 77843, United States
| | - Sarah E. Bondos
- Department of Molecular and Cellular Medicine, Texas A&M University Health Science Center, College Station, Texas 77843, United States
- Department
of Biochemistry and Cell Biology, Rice University, Houston, Texas 77005, United States
| | - Kayla J. Bayless
- Department of Molecular and Cellular Medicine, Texas A&M University Health Science Center, College Station, Texas 77843, United States
| |
Collapse
|
33
|
Schwarzlin R, Pušenjak N, Makuc D, Križman M, Vovk I, Plavec J, Švajger U. Synergistic complex from plants Solanaceae exhibits cytotoxicity for the human hepatocellular carcinoma cell line HepG2. Altern Ther Health Med 2016; 16:395. [PMID: 27756283 PMCID: PMC5070086 DOI: 10.1186/s12906-016-1362-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2016] [Accepted: 09/09/2016] [Indexed: 11/11/2022]
Abstract
Background It had been demonstrated that sugars from various plants can act as potent agents, which induce apoptosis of cancer cells. Methods Using HPLC, we fractionated a mixture of two plant extracts from the plant family Solanaceae, namely Capsicum chinense and the plant family Amaryllidaceae namely Allium sativum. We evaluated the effect of different fractions on apoptosis of HepG2 cell line. The most effective fraction was further studied to determine its molecular composition using mass spectrometry (MS) and NMR. We further evaluated the effect of determined molecular composition found in the selected fraction by using a mixture of commercially available substances, which were found in the fraction and tested its pro-apoptotic effect on HepG2 cells. To get some insight into potential apoptotic mechanisms we studied caspase-3 activity and mitochondrial integrity in treated cells. Results Out of 93 fractions obtained by HPLC from the plant extract we found HPLC fraction 10 (10 min elution) was the most effective. MS and NMR studies revealed high presence of cellobiose together with vitamin C, sulphur (S) and trace amounts of selenium (Se). HPLC fraction 10 triggered apoptosis of HepG2 within 3 h in the 0.01–1.0 mg/mL concentration range. Furthermore, a mixture of pure cellobiose, vitamin C, S and Se (complex cellobiose/C/S/Se) had a very similar capacity in inducing apoptosis of HepG2 cells compared to HPLC fraction 10. Complex cellobiose/C/S/Se was capable of inducing caspase-3 activity and led to loss of mitochondrial integrity. The capacity of cellobiose alone to induce apoptosis of HepG2 was approximately 1000-fold lower compared to complex cellobiose/C/S/Se. Conclusion In this study we present the highly synergistic effect of a unique complex consisting of cellobiose, vitamin C, sulphur and selenium on triggering the apoptosis of human hepatocellular carcinoma (HepG2) cell line.
Collapse
|
34
|
Abstract
AbstractThe endothelium, a thin single sheet of endothelial cells, is a metabolically active layer that coats the inner surface of blood vessels and acts as an interface between the circulating blood and the vessel wall. The endothelium through the secretion of vasodilators and vasoconstrictors serves as a critical mediator of vascular homeostasis. During the development of the vascular system, it regulates cellular adhesion and vessel wall inflammation in addition to maintaining vasculogenesis and angiogenesis. A shift in the functions of the endothelium towards vasoconstriction, proinflammatory and prothrombic states characterise improper functioning of these cells, leading to endothelial dysfunction (ED), implicated in the pathogenesis of many diseases including diabetes. Major mechanisms of ED include the down-regulation of endothelial nitric oxide synthase levels, differential expression of vascular endothelial growth factor, endoplasmic reticulum stress, inflammatory pathways and oxidative stress. ED tends to be the initial event in macrovascular complications such as coronary artery disease, peripheral arterial disease, stroke and microvascular complications such as nephropathy, neuropathy and retinopathy. Numerous strategies have been developed to protect endothelial cells against various stimuli, of which the role of polyphenolic compounds in modulating the differentially regulated pathways and thus maintaining vascular homeostasis has been proven to be beneficial. This review addresses the factors stimulating ED in diabetes and the molecular mechanisms of natural polyphenol antioxidants in maintaining vascular homeostasis.
Collapse
|
35
|
Fang F, Peng T, Yang S, Wang W, Zhang Y, Li H. Lycium barbarum
polysaccharide attenuates the cytotoxicity of mutant huntingtin and increases the activity of AKT. Int J Dev Neurosci 2016; 52:66-74. [DOI: 10.1016/j.ijdevneu.2016.05.004] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2016] [Revised: 05/02/2016] [Accepted: 05/02/2016] [Indexed: 01/13/2023] Open
Affiliation(s)
- Fang Fang
- Division of Histology and EmbryologyDepartment of AnatomyTongji Medical CollegeHuazhong University of Science and TechnologyWuhan430030People's Republic of China
| | - Ting Peng
- Division of Histology and EmbryologyDepartment of AnatomyTongji Medical CollegeHuazhong University of Science and TechnologyWuhan430030People's Republic of China
- Institute for Brain SciencesTongji Medical CollegeHuazhong University of Science and TechnologyWuhan430030People's Republic of China
- Collaborative Innovation Center for Brain ScienceHuazhong University of Science and TechnologyWuhan430030People's Republic of China
| | - Shiming Yang
- Division of Histology and EmbryologyDepartment of AnatomyTongji Medical CollegeHuazhong University of Science and TechnologyWuhan430030People's Republic of China
| | - Weixi Wang
- Division of Histology and EmbryologyDepartment of AnatomyTongji Medical CollegeHuazhong University of Science and TechnologyWuhan430030People's Republic of China
| | - Yinong Zhang
- Division of Histology and EmbryologyDepartment of AnatomyTongji Medical CollegeHuazhong University of Science and TechnologyWuhan430030People's Republic of China
| | - He Li
- Division of Histology and EmbryologyDepartment of AnatomyTongji Medical CollegeHuazhong University of Science and TechnologyWuhan430030People's Republic of China
- Institute for Brain SciencesTongji Medical CollegeHuazhong University of Science and TechnologyWuhan430030People's Republic of China
- Collaborative Innovation Center for Brain ScienceHuazhong University of Science and TechnologyWuhan430030People's Republic of China
| |
Collapse
|
36
|
Zaitseva II, Berggren PO, Zaitsev SV. Insulinotropic compounds decrease endothelial cell survival. Toxicol In Vitro 2016; 33:1-8. [PMID: 26883446 DOI: 10.1016/j.tiv.2016.02.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2015] [Revised: 01/05/2016] [Accepted: 02/10/2016] [Indexed: 10/22/2022]
Abstract
OBJECTIVES Hyperglycemia induces damage of vascular endothelial cells leading to diabetic complications. We investigated the effects of insulinotropic compounds and elevated glucose on endothelial cells in the absence or presence of vascular endothelial growth factor (VEGF). RESULTS Human umbilical vein endothelial cells (HUVECs) were treated with glibenclamide, repaglinide and insulinotropic imidazolines at high glucose concentration in the presence or absence of VEGF and viability, proliferation and nitric oxide production were measured. Hyperglycemia inhibited pro-survival effects of VEGF on endothelial cells. Glibenclamide and repaglinide decreased HUVEC viability at elevated glucose concentration in the absence but not in the presence of VEGF, without affecting HUVEC proliferation. Repaglinide also had some positive influence on HUVEC function elevating NO production in the presence of VEGF. Imidazolines showed different activities on endothelial cell survival. Efaroxan diminished HUVEC viability at elevated glucose concentration in the presence, however not in the absence of VEGF, while RX871024 decreased HUVEC survival regardless of the presence of VEGF. SIGNIFICANCE OF THE STUDY Our data demonstrate an important interplay between the actual insulinotropic compounds, VEGF and ambient glucose concentration affecting the survival of the vascular endothelial cells. Consequently, this interplay needs to be taken into consideration when designing novel oral antidiabetic compounds.
Collapse
Affiliation(s)
- Irina I Zaitseva
- Rolf Luft Research Center for Diabetes and Endocrinology, Karolinska Institutet, SE-17176 Stockholm, Sweden
| | - Per-Olof Berggren
- Rolf Luft Research Center for Diabetes and Endocrinology, Karolinska Institutet, SE-17176 Stockholm, Sweden
| | - Sergei V Zaitsev
- Rolf Luft Research Center for Diabetes and Endocrinology, Karolinska Institutet, SE-17176 Stockholm, Sweden; Lomonosov Moscow State University, Belozersky Institute of Physico-chemical Biology, Faculty of Bioengineering and Bioinformatics, Moscow 119992, Russia.
| |
Collapse
|
37
|
Sultan SA, Liu W, Peng Y, Roberts W, Whitelaw D, Graham AM. The Role of Maternal Gestational Diabetes in Inducing Fetal Endothelial Dysfunction. J Cell Physiol 2015; 230:2695-705. [PMID: 25808705 DOI: 10.1002/jcp.24993] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2014] [Accepted: 03/19/2015] [Indexed: 12/27/2022]
Abstract
Gestational diabetes mellitus (GDM) is known to be associated with fetal endothelial dysfunction, however, the mechanisms are not fully understood. This study examines the effect of maternal diabetes on fetal endothelial function and gene expression under physiological glucose conditions (5 mM). Human umbilical vein endothelial cell (HUVEC) isolated from diabetic mothers (d.HUVEC) grew more slowly than HUVEC isolated from healthy mothers (c.HUVEC) and had delayed doubling time despite increased levels of total vascular endothelial growth factor (VEGF) expression and protein production as determined by real-time PCR and ELISA respectively. Using western blot, the levels of antiproliferative VEGF165b isoform were increased in d.HUVEC relative to c.HUVEC. Successful VEGF165b knockdown by small interfering RNA (siRNA) resulted in increased proliferation of d.HUVEC measured by MTT, compared with negative siRNA control, to similar levels measured in c.HUVEC. In addition, d.HUVEC generated excess levels of ROS as revealed by 2',7' Dichlorodihydrofluorescein Diacetate (DCFH-DA) and Nitrotetrazolium blue (NBT). Using microarray, 102 genes were differentially overexpressed between d.HUVEC versus c.HUVEC (>1.5-fold change; P < 0.05). Functional clustering analysis of these differentially expressed genes revealed participation in inflammatory responses (including adhesion) which may be related to pathological outcomes. Of these genes, ICAM-1 was validated as upregulated, confirming microarray results. Additional confirmatory immunofluorescence staining revealed increased protein expression of ICAM-1 compared with c.HUVEC which was reduced by vitamin C treatment (100 μM). Thus, maternal diabetes induces persistent alterations in fetal endothelial function and gene expression following glucose normalization and antioxidant treatment could help reverse endothelium dysfunction.
Collapse
Affiliation(s)
- Samar A Sultan
- Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Wanting Liu
- School of Computing, Informatics and Media, University of Bradford, Bradford, UK
| | - Yonghong Peng
- School of Computing, Informatics and Media, University of Bradford, Bradford, UK
| | - W Roberts
- School of Medical Sciences, University of Bradford, Bradford, UK
| | - Donald Whitelaw
- Department of Diabetes and Endocrinology, Bradford Royal Infirmary, Bradford, UK
| | - Anne M Graham
- School of Medical Sciences, University of Bradford, Bradford, UK
| |
Collapse
|
38
|
Ouazia D, Levros LC, Rassart É, Desrosiers R. The protein l-isoaspartyl (d-aspartyl) methyltransferase protects against dopamine-induced apoptosis in neuroblastoma SH-SY5Y cells. Neuroscience 2015; 295:139-50. [DOI: 10.1016/j.neuroscience.2015.03.026] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2015] [Revised: 03/10/2015] [Accepted: 03/14/2015] [Indexed: 11/15/2022]
|
39
|
Kim S, Kwon J. Effect of thymosin beta 4 in the presence of up-regulation of the insulin-like growth factor-1 signaling pathway on high-glucose-exposed vascular endothelial cells. Mol Cell Endocrinol 2015; 401:238-47. [PMID: 25540921 DOI: 10.1016/j.mce.2014.12.008] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/01/2014] [Revised: 11/14/2014] [Accepted: 12/09/2014] [Indexed: 11/15/2022]
Abstract
Thymosin beta 4 (Tβ4), which regulates vascular cell growth, can ameliorate some of the problems associated with diabetes. However, the precise signaling mechanisms by which Tβ4 protects against hyperglycemia-induced damage to endothelial cells have not been investigated in detail. Thus, the aim of this study was to elucidate the role of Tβ4 in diabetes and the possible involvement of insulin-like growth factor-1 (IGF-1), which affects cellular survival, metabolism, and glucose homeostasis in high-glucose (HG)-injured human umbilical vein endothelial cells (HUVECs). Immunoblotting assays revealed that under HG blockade conditions, Tβ4 did not alter the insulin-signaling pathway, but induced overexpression of IGF-1 protein, leading to activation of factors in alternative signaling pathway. Small interfering RNA of Tβ4 and IGF-1 were studied to clarify relationship between Tβ4 and IGF-1. These findings suggest that IGF-1 induction by Tβ4 ameliorates the damage in HG-injured HUVECs which manifest as diabetic vascular disorder.
Collapse
Affiliation(s)
- Sokho Kim
- Department of Laboratory Animal Medicine, College of Veterinary Medicine, Chonbuk National University, Jeonju, Jeonbuk 561-156, Republic of Korea
| | - Jungkee Kwon
- Department of Laboratory Animal Medicine, College of Veterinary Medicine, Chonbuk National University, Jeonju, Jeonbuk 561-156, Republic of Korea.
| |
Collapse
|
40
|
Critical Role of Prohibitin in Endothelial Cell Apoptosis Caused by Glycated Low-density Lipoproteins and Protective Effects of Grape Seed Procyanidin B2. J Cardiovasc Pharmacol 2015; 65:13-21. [DOI: 10.1097/fjc.0000000000000157] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
41
|
Abstract
For years, diabetic retinopathy has been defined based on vascular lesions, and neural abnormalities were not regarded as important. This review summarizes evidence that the neural retina has important effects on the retinal vasculature under normal conditions, and the interaction between the retinal neuroglial cells and vascular function is altered in diabetes. Importantly, new evidence raises a possibility that abnormalities within retinal neuroglial cells (notably photoreceptors) might actually be causing or initiating the vascular disease in diabetic retinopathy.
Collapse
Affiliation(s)
- Timothy S. Kern
- Department of Medicine, Case Western Reserve University Medicine, Cleveland, OH, USA
- Department of Medicine, Louis Stokes Cleveland Veterans Administration Medical Center, Cleveland, OH, USA
| |
Collapse
|
42
|
Shao B, Bayraktutan U. Hyperglycaemia promotes human brain microvascular endothelial cell apoptosis via induction of protein kinase C-ßI and prooxidant enzyme NADPH oxidase. Redox Biol 2014; 2:694-701. [PMID: 24936444 PMCID: PMC4052534 DOI: 10.1016/j.redox.2014.05.005] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2014] [Revised: 05/21/2014] [Accepted: 05/23/2014] [Indexed: 01/13/2023] Open
Abstract
Blood-brain barrier disruption represents a key feature in hyperglycaemia-aggravated cerebral damage after an ischaemic stroke. Although the underlying mechanisms remain largely unknown, activation of protein kinase C (PKC) is thought to play a critical role. This study examined whether apoptosis of human brain microvascular endothelial cells (HBMEC) might contribute to hyperglycaemia-evoked barrier damage and assessed the specific role of PKC in this phenomenon. Treatments with hyperglycaemia (25 mM) or phorbol myristate acetate (PMA, a protein kinase C activator, 100 nM) significantly increased NADPH oxidase activity, O2 (•-) generation, proapoptotic protein Bax expression, TUNEL-positive staining and caspase-3/7 activities. Pharmacological inhibition of NADPH oxidase, PKC-a, PKC-ß or PKC-ßI via their specific inhibitors and neutralisation of O2 (•-) by a cell-permeable superoxide dismutase mimetic, MnTBAP normalised all the aforementioned increases induced by hyperglycaemia. Suppression of these PKC isoforms also negated the stimulatory effects of hyperglycaemia on the protein expression of NADPH oxidase membrane-bound components, Nox2 and p22-phox which determine the overall enzymatic activity. Silencing of PKC-ßI gene through use of specific siRNAs abolished the effects of both hyperglycaemia and PMA on endothelial cell NADPH oxidase activity, O2 (•-) production and apoptosis and consequently improved the integrity and function of an in vitro model of human cerebral barrier comprising HBMEC, astrocytes and pericytes. Hyperglycaemia-mediated apoptosis of HBMEC contributes to cerebral barrier dysfunction and is modulated by sequential activations of PKC-ßI and NADPH oxidase.
Collapse
Affiliation(s)
- Beili Shao
- Stroke, Division of Clinical Neuroscience, School of Medicine, University of Nottingham, UK
| | - Ulvi Bayraktutan
- Stroke, Division of Clinical Neuroscience, School of Medicine, University of Nottingham, UK
| |
Collapse
|
43
|
Abbasi N, Akhavan MM, Rahbar-Roshandel N, Shafiei M. The effects of low and high concentrations of luteolin on cultured human endothelial cells under normal and glucotoxic conditions: involvement of integrin-linked kinase and cyclooxygenase-2. Phytother Res 2014; 28:1301-7. [PMID: 25201753 DOI: 10.1002/ptr.5128] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2013] [Revised: 01/04/2014] [Accepted: 01/14/2014] [Indexed: 12/12/2022]
Abstract
Luteolin protects against high glucose (HG)-induced endothelial dysfunction whereas its cytotoxicity has been reported against normal endothelial cells. This study was undertaken to determine luteolin cytoprotective and cytotoxic dose ranges and to elucidate their respective mechanisms. Luteolin prevented HG-induced human umbilical vein endothelial cell (HUVEC) death with an EC50 value of 2.0 ± 0.07 μM. The protective effect of luteolin was associated with decreased intracellular reactive oxygen species (ROS) and Ca(2+) (Cai(2+)) levels and enhanced nitric oxide (NO) production. At high concentrations, luteolin caused HUVEC death in normal glucose (NG) and HG states (LC50 40 ± 2.23 and 38 ± 1.12 μM, respectively), as represented by increased ROS and Cai(2+) and decreased NO. Western blots illustrated that exposure to HG increased cyclooxygenase-2 (COX-2) and integrin-linked kinase (ILK) expression. Luteolin at low concentrations suppressed HG-mediated up-regulation of COX-2 but maintained HG-induced over-expression of ILK while at high concentrations significantly increased COX-2 and decreased ILK expression in both HG and NG states. Our data indicated that cytoprotective action of luteolin was manifested with much lower concentrations, by a factor of approximately 20, compared with cytotoxic activity under both normal or glucotoxic conditions. It appears that luteolin exerts its action, in part, by modulating ILK expression which is associated with regulation of COX-2 expression and NO production in endothelial cells.
Collapse
Affiliation(s)
- Naser Abbasi
- Department of Pharmacology, Medical School, Iran University of Medical Sciences, Tehran, Iran
| | | | | | | |
Collapse
|
44
|
Dunn LL, Simpson PJ, Prosser HC, Lecce L, Yuen GS, Buckle A, Sieveking DP, Vanags LZ, Lim PR, Chow RW, Lam YT, Clayton Z, Bao S, Davies MJ, Stadler N, Celermajer DS, Stocker R, Bursill CA, Cooke JP, Ng MK. A critical role for thioredoxin-interacting protein in diabetes-related impairment of angiogenesis. Diabetes 2014; 63:675-87. [PMID: 24198286 PMCID: PMC3900553 DOI: 10.2337/db13-0417] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Impaired angiogenesis in ischemic tissue is a hallmark of diabetes. Thioredoxin-interacting protein (TXNIP) is an exquisitely glucose-sensitive gene that is overexpressed in diabetes. As TXNIP modulates the activity of the key angiogenic cytokine vascular endothelial growth factor (VEGF), we hypothesized that hyperglycemia-induced dysregulation of TXNIP may play a role in the pathogenesis of impaired angiogenesis in diabetes. In the current study, we report that high glucose-mediated overexpression of TXNIP induces a widespread impairment in endothelial cell (EC) function and survival by reducing VEGF production and sensitivity to VEGF action, findings that are rescued by silencing TXNIP with small interfering RNA. High glucose-induced EC dysfunction was recapitulated in normal glucose conditions by overexpressing either TXNIP or a TXNIP C247S mutant unable to bind thioredoxin, suggesting that TXNIP effects are largely independent of thioredoxin activity. In streptozotocin-induced diabetic mice, TXNIP knockdown to nondiabetic levels rescued diabetes-related impairment of angiogenesis, arteriogenesis, blood flow, and functional recovery in an ischemic hindlimb. These findings were associated with in vivo restoration of VEGF production to nondiabetic levels. These data implicate a critical role for TXNIP in diabetes-related impairment of ischemia-mediated angiogenesis and identify TXNIP as a potential therapeutic target for the vascular complications of diabetes.
Collapse
Affiliation(s)
- Louise L. Dunn
- Translational Research Group, The Heart Research Institute, Sydney, Australia
- Sydney Medical School, University of Sydney, Sydney, Australia
| | | | - Hamish C. Prosser
- Immunobiology Group, The Heart Research Institute, Sydney, Australia
| | - Laura Lecce
- Translational Research Group, The Heart Research Institute, Sydney, Australia
| | - Gloria S.C. Yuen
- Translational Research Group, The Heart Research Institute, Sydney, Australia
- Sydney Medical School, University of Sydney, Sydney, Australia
| | - Andrew Buckle
- Translational Research Group, The Heart Research Institute, Sydney, Australia
| | - Daniel P. Sieveking
- Translational Research Group, The Heart Research Institute, Sydney, Australia
| | - Laura Z. Vanags
- Immunobiology Group, The Heart Research Institute, Sydney, Australia
| | - Patrick R. Lim
- Translational Research Group, The Heart Research Institute, Sydney, Australia
| | - Renee W.Y. Chow
- Translational Research Group, The Heart Research Institute, Sydney, Australia
| | - Yuen Ting Lam
- Translational Research Group, The Heart Research Institute, Sydney, Australia
| | - Zoe Clayton
- Translational Research Group, The Heart Research Institute, Sydney, Australia
| | - Shisan Bao
- Sydney Medical School, University of Sydney, Sydney, Australia
| | - Michael J. Davies
- Sydney Medical School, University of Sydney, Sydney, Australia
- Free Radical Group, The Heart Research Institute, Sydney, Australia
| | - Nadina Stadler
- Free Radical Group, The Heart Research Institute, Sydney, Australia
| | - David S. Celermajer
- Sydney Medical School, University of Sydney, Sydney, Australia
- Clinical Research Group, The Heart Research Institute, Sydney, Australia
- Department of Cardiology, Royal Prince Alfred Hospital, Sydney, Australia
| | - Roland Stocker
- Vascular Biology Division, Victor Chang Cardiac Research Institute, Sydney, Australia
| | | | - John P. Cooke
- Department of Medicine, Stanford University School of Medicine, Stanford, CA
| | - Martin K.C. Ng
- Translational Research Group, The Heart Research Institute, Sydney, Australia
- Sydney Medical School, University of Sydney, Sydney, Australia
- Department of Cardiology, Royal Prince Alfred Hospital, Sydney, Australia
- Corresponding author: Martin K.C. Ng,
| |
Collapse
|
45
|
Patel H, Chen J, Das KC, Kavdia M. Hyperglycemia induces differential change in oxidative stress at gene expression and functional levels in HUVEC and HMVEC. Cardiovasc Diabetol 2013; 12:142. [PMID: 24093550 PMCID: PMC3851327 DOI: 10.1186/1475-2840-12-142] [Citation(s) in RCA: 124] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2013] [Accepted: 10/04/2013] [Indexed: 01/14/2023] Open
Abstract
Background Endothelial dysfunction precedes pathogenesis of vascular complications in diabetes. In recent years, the mechanisms of endothelial dysfunction were investigated to outline strategies for its treatment. However, the therapies for dysfunctional endothelium resulted in multiple clinical trial failures and remain elusive. There is a need for defining hyperglycemia-induced endothelial dysfunction with both generic and specific dysfunctional changes in endothelial cells (EC) using a systems approach. In this study, we investigated hyperglycemia-induced endothelial dysfunction in HUVEC and HMVEC. We investigated hyperglycemia-induced functional changes (superoxide (O2‾), and hydrogen peroxide (H2O2) production and mitochondrial membrane polarization) and gene expression fingerprints of related enzymes (nitric oxide synthase, NAD(P)H oxidase, and reactive oxygen species (ROS) neutralizing enzymes) in both ECs. Method Gene expression of NOS2, NOS3, NOX4, CYBA, UCP1, CAT, TXNRD1, TXNRD2, GPX1, NOX1, SOD1, SOD2, PRDX1, 18s, and RPLP0 were measured using real-time PCR. O2‾ production was measured with dihydroethidium (DHE) fluorescence measurement. H2O2 production was measured using Amplex Red assay. Mitochondrial membrane polarization was measured using JC-10 based fluorescence measurement. Results We showed that the O2‾ levels increased similarly in both ECs with hyperglycemia. However, these endothelial cells showed significantly different underlying gene expression profile, H2O2 production and mitochondrial membrane polarization. In HUVEC, hyperglycemia increased H2O2 production, and hyperpolarized mitochondrial membrane. ROS neutralizing enzymes SOD2 and CAT gene expression were downregulated. In contrast, there was an upregulation of nitric oxide synthase and NAD(P)H oxidase and a depolarization of mitochondrial membrane in HMVEC. In addition, ROS neutralizing enzymes SOD1, GPX1, TXNRD1 and TXNRD2 gene expression were significantly upregulated in high glucose treated HMVEC. Conclusion Our findings highlighted a unique framework for hyperglycemia-induced endothelial dysfunction. We showed that multiple pathways are differentially affected in these endothelial cells in hyperglycemia. High occurrences of gene expression changes in HMVEC in this study supports the hypothesis that microvasculature precedes macrovasculature in epigenetic regulation forming vascular metabolic memory. Identifying genomic phenotype and corresponding functional changes in hyperglycemic endothelial dysfunction will provide a suitable systems biology approach for understanding underlying mechanisms and possible effective therapeutic intervention.
Collapse
Affiliation(s)
- Hemang Patel
- Department of Biomedical Engineering, Wayne State University, 2322 Engineering, 5050 Anthony Wayne Dr,, Detroit, MI 48202, USA.
| | | | | | | |
Collapse
|
46
|
Li XL, Li BY, Cheng M, Yu F, Yin WB, Cai Q, Zhang Z, Zhang JH, Wang JF, Zhou RH, Gao HQ. PIMT prevents the apoptosis of endothelial cells in response to glycated low density lipoproteins and protective effects of grape seed procyanidin B2. PLoS One 2013; 8:e69979. [PMID: 23922881 PMCID: PMC3724603 DOI: 10.1371/journal.pone.0069979] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2013] [Accepted: 06/14/2013] [Indexed: 11/28/2022] Open
Abstract
Background The development of diabetic angiopathy is associated with profound vascular endothelial cells (VEC) dysfunction and apoptosis. Glycated low density lipoproteins (gly-LDL) continuously produced in the setting of diabetic patients play an important role in causing VEC dysfunction and apoptosis. However, the underlying molecular mechanism remains largely elusive. Protein L-isoaspartyl methyltransferase (PIMT) is a widely expressed protein repair enzyme by multiple cell types of arterial wall including VEC. Our previous proteomic studies showed that the expression of PIMT was significantly decreased in the aorta of diabetic rats as compared with control rats and treatment with grape seed procyanidin extracts significantly increased the PIMT expression in diabetic rats. We hypothesized that PIMT plays a critical role in gly-LDL induced VEC apoptosis; grape seed procyanidin B2 (GSPB2) protect against gly-LDL induced VEC apoptosis through PIMT regulation. Methods and Results HUVEC transfected negative control and PIMT siRNA were treated with or without GSPB2 (10 µmol/L) for 48 h. Moreover, HUVEC of PIMT overexpression were stimulated by gly-LDL (50 µg/ml) in the presence or absence of GSPB2 (10 µmol/L) for 48 h. Our results showed that gly-LDL downregulated PIMT expression and PIMT overexpression or GSPB2 significantly attenuated gly-LDL induced VEC apoptosis. PIMT siRNA increased VEC apoptosis with up-regulation of p53, cytochrome c release, caspase-9 and caspase-3 activation. Mechanistically, overexpression of PIMT or GSPB2 increased the phosphorylation of ERK1/2 and GSK3β in the gly-LDL induced VEC. Conclusion In summary, our study identified PIMT as a key player responsible for gly-LDL induced VEC apoptosis and GSPB2 protect against gly-LDL induced VEC apoptosis by PIMT up-regulation. Targeting PIMT including use of GSPB2 could be turned into clinical application in the fighting against diabetic vascular complications.
Collapse
Affiliation(s)
- Xiao-li Li
- Key Laboratory of Cardiovascular Proteomics of Shandong Province, Department of Geriatric Endocrinology, Qi-Lu Hospital of Shandong University, Jinan, China
- Department of Drug Purchase and Supply, Qi-Lu Hospital of Shandong University, Jinan, China
| | - Bao-ying Li
- Key Laboratory of Cardiovascular Proteomics of Shandong Province, Department of Geriatric Endocrinology, Qi-Lu Hospital of Shandong University, Jinan, China
| | - Mei Cheng
- Key Laboratory of Cardiovascular Proteomics of Shandong Province, Department of Geriatric Endocrinology, Qi-Lu Hospital of Shandong University, Jinan, China
| | - Fei Yu
- Key Laboratory of Cardiovascular Proteomics of Shandong Province, Department of Geriatric Endocrinology, Qi-Lu Hospital of Shandong University, Jinan, China
| | - Wen-bin Yin
- Key Laboratory of Cardiovascular Proteomics of Shandong Province, Department of Geriatric Endocrinology, Qi-Lu Hospital of Shandong University, Jinan, China
| | - Qian Cai
- Key Laboratory of Cardiovascular Proteomics of Shandong Province, Department of Geriatric Endocrinology, Qi-Lu Hospital of Shandong University, Jinan, China
| | - Zhen Zhang
- Key Laboratory of Cardiovascular Proteomics of Shandong Province, Department of Geriatric Endocrinology, Qi-Lu Hospital of Shandong University, Jinan, China
| | - Jian-hua Zhang
- Institute of Basic Science, Medical Science Academy of Shandong, Jinan, China
| | - Jun-fu Wang
- Institute of Basic Science, Medical Science Academy of Shandong, Jinan, China
| | - Rui-hai Zhou
- Division of Cardiology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
| | - Hai-qing Gao
- Key Laboratory of Cardiovascular Proteomics of Shandong Province, Department of Geriatric Endocrinology, Qi-Lu Hospital of Shandong University, Jinan, China
- * E-mail:
| |
Collapse
|
47
|
Pangare M, Makino A. Mitochondrial function in vascular endothelial cell in diabetes. J Smooth Muscle Res 2012; 48:1-26. [PMID: 22504486 DOI: 10.1540/jsmr.48.1] [Citation(s) in RCA: 67] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
Micro- and macrovascular complications are commonly seen in diabetic patients and endothelial dysfunction contributes to the development and progression of the complications. Abnormal functions in endothelial cells lead to the increase in vascular tension and atherosclerosis, followed by systemic hypertension as well as increased incidence of ischemia and stroke in diabetic patients. Mitochondria are organelles serving as a source of energy production and as regulators of cell survival (e.g., apoptosis and cell development) and ion homeostasis (e.g., H(+), Ca(2+)). Endothelial mitochondria are mainly responsible for generation of reactive oxygen species (ROS) and maintaining the Ca(2+) concentration in the cytosol. There is increasing evidence that mitochondrial morphological and functional changes are implicated in vascular endothelial dysfunction. Enhanced mitochondrial fission and/or attenuated fusion lead to mitochondrial fragmentation and disrupt the endothelial physiological function. Abnormal mitochondrial biogenesis and disturbance of mitochondrial autophagy increase the accumulation of damaged mitochondria, such as irreversibly depolarized or leaky mitochondria, and facilitate cell death. Augmented mitochondrial ROS production and Ca(2+) overload in mitochondria not only cause the maladaptive effect on the endothelial function, but also are potentially detrimental to cell survival. In this article, we review the physiological and pathophysiological role of mitochondria in endothelial function with special focus on diabetes.
Collapse
Affiliation(s)
- Meenal Pangare
- University of Illinois at Chicago, Chicago, IL 60612, USA
| | | |
Collapse
|
48
|
Karbach S, Jansen T, Horke S, Heeren T, Scholz A, Coldewey M, Karpi A, Hausding M, Kröller-Schön S, Oelze M, Münzel T, Daiber A. Hyperglycemia and oxidative stress in cultured endothelial cells--a comparison of primary endothelial cells with an immortalized endothelial cell line. J Diabetes Complications 2012; 26:155-62. [PMID: 22521318 DOI: 10.1016/j.jdiacomp.2012.03.011] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/24/2012] [Revised: 02/29/2012] [Accepted: 03/01/2012] [Indexed: 01/01/2023]
Abstract
Diabetes mellitus is a major risk factor for the development of cardiovascular disease and oxidative stress plays an important role in this process. Therefore, we investigated the effects of hyperglycemia on the formation of reactive oxygen species (ROS) and nitric oxide/cGMP signaling in two different endothelial cell cultures. Human umbilical vein endothelial cells (HUVEC) and EA.hy 926 cells showed increased oxidative stress and impaired NO-cGMP signaling in response to hyperglycemia. The major difference between the two different cell types was the dramatic decrease in viability in HUVEC whereas EA.hy cells showed rather increased growth under hyperglycemic conditions. Starvation led to an additional substantial decrease in viability and increased superoxide formation in HUVEC. Both endothelial cell types, HUVEC and EA.hy 926, may be used as models for vascular hyperglycemia. However, high growth medium should be used to avoid starvation-induced oxidative stress and cell death.
Collapse
Affiliation(s)
- Susanne Karbach
- 2nd Medical Clinic, Department of Cardiology, Medical Center of the Johannes Gutenberg University, Mainz, Germany
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Keats EC, Khan ZA. Vascular stem cells in diabetic complications: evidence for a role in the pathogenesis and the therapeutic promise. Cardiovasc Diabetol 2012; 11:37. [PMID: 22524626 PMCID: PMC3476432 DOI: 10.1186/1475-2840-11-37] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/07/2012] [Accepted: 04/23/2012] [Indexed: 12/25/2022] Open
Abstract
Long standing diabetes leads to structural and functional alterations in both the micro- and the macro-vasculature. Vascular endothelial cells (ECs) are the primary target of the hyperglycemia-induced adverse effects. Vascular stem cells that give rise to endothelial progenitor cells (EPCs) and mesenchymal progenitor cells (MPCs) represent an attractive target for cell therapy for diabetic patients. A number of studies have reported EPC dysfunction as a novel participant in the culmination of the diabetic complications. The controversy behind the identity of EPCs and the similarity between these progenitor cells to hematopoietic cells has led to conflicting results. MPCs, on the other hand, have not been examined for a potential role in the pathogenesis of the complications. These multipotent cells, however, do show a therapeutic role. In this article, we summarize the vascular changes that occur in diabetic complications highlighting some of the common features, the key findings that illustrate an important role of vascular stem cells (VSCs) in the pathogenesis of chronic diabetic complications, and provide mechanisms by which these cells can be used for therapy.
Collapse
MESH Headings
- Animals
- Blood Glucose/metabolism
- Diabetic Angiopathies/blood
- Diabetic Angiopathies/pathology
- Diabetic Angiopathies/physiopathology
- Diabetic Angiopathies/surgery
- Endothelial Cells/metabolism
- Endothelial Cells/pathology
- Endothelial Cells/transplantation
- Endothelium, Vascular/metabolism
- Endothelium, Vascular/pathology
- Endothelium, Vascular/physiopathology
- Humans
- Mesenchymal Stem Cell Transplantation
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/pathology
- Muscle, Smooth, Vascular/physiopathology
- Neovascularization, Physiologic
- Regeneration
- Treatment Outcome
Collapse
Affiliation(s)
- Emily C Keats
- Department of Pathology, University of Western Ontario, London, ON, Canada
| | - Zia A Khan
- Department of Pathology, University of Western Ontario, London, ON, Canada
- Metabolism and Diabetes Program, Lawson Health Research Institute, London, ON, Canada
- 4011 Dental Sciences Building, 1151 Richmond Street, London, ON, N6A 5C1, Canada
| |
Collapse
|
50
|
Gu X, Yao Y, Cheng R, Zhang Y, Dai Z, Wan G, Yang Z, Cai W, Gao G, Yang X. Plasminogen K5 activates mitochondrial apoptosis pathway in endothelial cells by regulating Bak and Bcl-x(L) subcellular distribution. Apoptosis 2011; 16:846-55. [PMID: 21656147 DOI: 10.1007/s10495-011-0618-9] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Plasminogen Kringle 5(K5) is a proteolytic fragment of plasminogen, which displays potent anti-angiogenic activities. K5 has been shown to induce apoptosis in proliferating endothelial cells; however the exact mechanism has not been well explored. The present study was designed to elucidate the possible molecular mechanism of K5-induced endothelial cell apoptosis. Our results showed that K5 inhibited basic fibroblast growth factors activated in human umbilical vein endothelial cells (HUVECs), indicating proliferation in a dose-dependent manner and induced endothelial cell death via apoptosis. K5 exposure activated caspase 7, 8 and 9. These results suggested that both the intrinsic mitochondrial apoptosis pathway and extrinsic pathway might be involved in K5-induced apoptosis. K5 reduced mitochondrial membrane potential (MMP) of HUVECs, demonstrating mitochondrial depolarization in HUVECs. K5 increased the ratio of Bak to Bcl-x(L) on mitochondria, decreased the ratio in cytosol, and had no effect on the total amounts of these proteins. K5 also did not effect on Bax/Bcl-2 distribution. K5 increased the ratio of Bak to Bcl-x(L) on mitochondrial that resulted in mitochondrial depolarization, cytochrome c release and consequently the cleavage of caspase 9. These results suggested that K5 induces endothelial cell apoptosis at least in part via activating mitochondrial apoptosis pathway. The regulation of K5 on Bak and Bcl-x(L) distribution may play an important role in endothelial cell apoptosis. These results provide further insight into the anti-angiogenesis roles of K5 in angiogenesis-related ocular diseases and solid tumors.
Collapse
Affiliation(s)
- Xiaoqiong Gu
- Department of Biochemistry, Zhongshan Medical School, Sun Yat-sen University, Guangzhou 510080, Guangdong Province, China
| | | | | | | | | | | | | | | | | | | |
Collapse
|