1
|
Pandian K, van Zonneveld AJ, Harms A, Hankemeier T. Metabolic alterations of endothelial cells under transient and persistent hypoxia: study using a 3D microvessels-on-chip model. Tissue Barriers 2024:2431416. [PMID: 39584359 DOI: 10.1080/21688370.2024.2431416] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Revised: 11/08/2024] [Accepted: 11/12/2024] [Indexed: 11/26/2024] Open
Abstract
Numerous signaling pathways are activated during hypoxia to facilitate angiogenesis, promoting interactions among endothelial cells and initiating downstream signaling cascades. Although the pivotal role of the nitric oxide (NO) response pathway is well-established, the involvement of arginine-specific metabolism and bioactive lipid mechanisms in 3D flow-activated in vitro models remains less understood. In this study, we explored the levels of arginine-specific metabolites and bioactive lipids in human coronary artery endothelial cells (HCAECs) under both transient and persistent hypoxia. We compared targeted metabolite levels between a 2D static culture model and a 3D microvessels-on-chip model. Notably, we observed robust regulation of NO metabolites in both transient and persistent hypoxic conditions. In the 2D model under transient hypoxia, metabolic readouts of bioactive lipids revealed increased oxidative stress markers, a phenomenon not observed in the 3D microvessels. Furthermore, we made a novel discovery that the responses of bioactive lipids were regulated by hypoxia inducible factor-1α (HIF-1α) in the 2D cell culture model and partially by HIF-1α and flow-induced shear stress in the 3D microvessels. Immunostaining confirmed the HIF-1α-induced regulation under both hypoxic conditions. Real-time oxygen measurements in the 3D microvessels using an oxygen probe validated that oxygen levels were maintained in the 3D model. Overall, our findings underscore the critical regulatory roles of HIF-1α and shear stress in NO metabolites and bioactive lipids in both 2D and 3D cell culture models.
Collapse
Affiliation(s)
- Kanchana Pandian
- Division of Systems Biomedicine and Pharmacology, LACDR, Leiden University, Leiden, The Netherlands
| | - Anton Jan van Zonneveld
- Department of Internal Medicine (Nephrology) and the Einthoven Laboratory for Vascular and Regenerative Medicine, Leiden University Medical Center (LUMC), Leiden, The Netherlands
| | - Amy Harms
- Division of Systems Biomedicine and Pharmacology, LACDR, Leiden University, Leiden, The Netherlands
| | - Thomas Hankemeier
- Division of Systems Biomedicine and Pharmacology, LACDR, Leiden University, Leiden, The Netherlands
| |
Collapse
|
2
|
Park S, Cho SW. Bioengineering toolkits for potentiating organoid therapeutics. Adv Drug Deliv Rev 2024; 208:115238. [PMID: 38447933 DOI: 10.1016/j.addr.2024.115238] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Revised: 01/28/2024] [Accepted: 02/27/2024] [Indexed: 03/08/2024]
Abstract
Organoids are three-dimensional, multicellular constructs that recapitulate the structural and functional features of specific organs. Because of these characteristics, organoids have been widely applied in biomedical research in recent decades. Remarkable advancements in organoid technology have positioned them as promising candidates for regenerative medicine. However, current organoids still have limitations, such as the absence of internal vasculature, limited functionality, and a small size that is not commensurate with that of actual organs. These limitations hinder their survival and regenerative effects after transplantation. Another significant concern is the reliance on mouse tumor-derived matrix in organoid culture, which is unsuitable for clinical translation due to its tumor origin and safety issues. Therefore, our aim is to describe engineering strategies and alternative biocompatible materials that can facilitate the practical applications of organoids in regenerative medicine. Furthermore, we highlight meaningful progress in organoid transplantation, with a particular emphasis on the functional restoration of various organs.
Collapse
Affiliation(s)
- Sewon Park
- Department of Biotechnology, Yonsei University, Seoul 03722, Republic of Korea
| | - Seung-Woo Cho
- Department of Biotechnology, Yonsei University, Seoul 03722, Republic of Korea; Center for Nanomedicine, Institute for Basic Science (IBS), Seoul 03722, Republic of Korea; Graduate Program of Nano Biomedical Engineering (NanoBME), Advanced Science Institute, Yonsei University, Seoul 03722, Republic of Korea.
| |
Collapse
|
3
|
Ye L, Liu R, Li Q, Zhou C, Tan X. Dysregulated VEGF/VEGFR-2 Signaling and Plexogenic Lesions in the Embryonic Lungs of Chickens Predisposed to Pulmonary Arterial Hypertension. Int J Mol Sci 2024; 25:4489. [PMID: 38674074 PMCID: PMC11049811 DOI: 10.3390/ijms25084489] [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: 12/13/2023] [Revised: 04/03/2024] [Accepted: 04/12/2024] [Indexed: 04/28/2024] Open
Abstract
Plexiform lesions are a hallmark of pulmonary arterial hypertension (PAH) in humans and are proposed to stem from dysfunctional angioblasts. Broiler chickens (Gallus gallus) are highly susceptible to PAH, with plexiform-like lesions observed in newly hatched individuals. Here, we reported the emergence of plexiform-like lesions in the embryonic lungs of broiler chickens. Lung samples were collected from broiler chickens at embryonic day 20 (E20), hatch, and one-day-old, with PAH-resistant layer chickens as controls. Plexiform lesions consisting of CD133+/vascular endothelial growth factor receptor type-2 (VEGFR-2)+ angioblasts were exclusively observed in broiler embryos and sporadically in layer embryos. Distinct gene profiles of angiogenic factors were observed between the two strains, with impaired VEGF-A/VEGFR-2 signaling correlating with lesion development and reduced arteriogenesis. Pharmaceutical inhibition of VEGFR-2 resulted in enhanced lesion development in layer embryos. Moreover, broiler embryonic lungs displayed increased activation of HIF-1α and nuclear factor erythroid 2-related factor 2 (Nrf2), indicating a hypoxic state. Remarkably, we found a negative correlation between lung Nrf2 activation and VEGF-A and VEGFR-2 expression. In vitro studies indicated that Nrf2 overactivation restricted VEGF signaling in endothelial progenitor cells. The findings from broiler embryos suggest an association between plexiform lesion development and impaired VEGF system due to aberrant activation of Nrf2.
Collapse
Affiliation(s)
- Lujie Ye
- Department of Veterinary Medicine, Zhejiang University, Hangzhou 310058, China
- Center for Veterinary Sciences, Zhejiang University, Hangzhou 310058, China
| | - Rui Liu
- Department of Veterinary Medicine, Zhejiang University, Hangzhou 310058, China
- Center for Veterinary Sciences, Zhejiang University, Hangzhou 310058, China
| | - Qinghao Li
- Department of Veterinary Medicine, Zhejiang University, Hangzhou 310058, China
- Center for Veterinary Sciences, Zhejiang University, Hangzhou 310058, China
| | - Chunzhen Zhou
- Department of Veterinary Medicine, Zhejiang University, Hangzhou 310058, China
- Center for Veterinary Sciences, Zhejiang University, Hangzhou 310058, China
| | - Xun Tan
- Department of Veterinary Medicine, Zhejiang University, Hangzhou 310058, China
- Center for Veterinary Sciences, Zhejiang University, Hangzhou 310058, China
- Institute of Preventive Veterinary Sciences, Zhejiang University, Hangzhou 310058, China
| |
Collapse
|
4
|
Oliveira RHDM, Annex BH, Popel AS. Endothelial cells signaling and patterning under hypoxia: a mechanistic integrative computational model including the Notch-Dll4 pathway. Front Physiol 2024; 15:1351753. [PMID: 38455844 PMCID: PMC10917925 DOI: 10.3389/fphys.2024.1351753] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Accepted: 02/12/2024] [Indexed: 03/09/2024] Open
Abstract
Introduction: Several signaling pathways are activated during hypoxia to promote angiogenesis, leading to endothelial cell patterning, interaction, and downstream signaling. Understanding the mechanistic signaling differences between endothelial cells under normoxia and hypoxia and their response to different stimuli can guide therapies to modulate angiogenesis. We present a novel mechanistic model of interacting endothelial cells, including the main pathways involved in angiogenesis. Methods: We calibrate and fit the model parameters based on well-established modeling techniques that include structural and practical parameter identifiability, uncertainty quantification, and global sensitivity. Results: Our results indicate that the main pathways involved in patterning tip and stalk endothelial cells under hypoxia differ, and the time under hypoxia interferes with how different stimuli affect patterning. Additionally, our simulations indicate that Notch signaling might regulate vascular permeability and establish different Nitric Oxide release patterns for tip/stalk cells. Following simulations with various stimuli, our model suggests that factors such as time under hypoxia and oxygen availability must be considered for EC pattern control. Discussion: This project provides insights into the signaling and patterning of endothelial cells under various oxygen levels and stimulation by VEGFA and is our first integrative approach toward achieving EC control as a method for improving angiogenesis. Overall, our model provides a computational framework that can be built on to test angiogenesis-related therapies by modulation of different pathways, such as the Notch pathway.
Collapse
Affiliation(s)
| | - Brian H. Annex
- Medical College of Georgia, Augusta University, Augusta, GA, United States
| | - Aleksander S. Popel
- Department of Biomedical Engineering, School of Medicine, Johns Hopkins University, Baltimore, MD, United States
| |
Collapse
|
5
|
Sivagurunathan N, Calivarathan L. SARS-CoV-2 Infection to Premature Neuronal Aging and Neurodegenerative Diseases: Is there any Connection with Hypoxia? CNS & NEUROLOGICAL DISORDERS DRUG TARGETS 2024; 23:431-448. [PMID: 37073650 DOI: 10.2174/1871527322666230418114446] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/26/2022] [Revised: 01/28/2023] [Accepted: 02/09/2023] [Indexed: 04/20/2023]
Abstract
The pandemic of coronavirus disease-2019 (COVID-19), caused by SARS-CoV-2, has become a global concern as it leads to a spectrum of mild to severe symptoms and increases death tolls around the world. Severe COVID-19 results in acute respiratory distress syndrome, hypoxia, and multi- organ dysfunction. However, the long-term effects of post-COVID-19 infection are still unknown. Based on the emerging evidence, there is a high possibility that COVID-19 infection accelerates premature neuronal aging and increases the risk of age-related neurodegenerative diseases in mild to severely infected patients during the post-COVID period. Several studies correlate COVID-19 infection with neuronal effects, though the mechanism through which they contribute to the aggravation of neuroinflammation and neurodegeneration is still under investigation. SARS-CoV-2 predominantly targets pulmonary tissues and interferes with gas exchange, leading to systemic hypoxia. The neurons in the brain require a constant supply of oxygen for their proper functioning, suggesting that they are more vulnerable to any alteration in oxygen saturation level that results in neuronal injury with or without neuroinflammation. We hypothesize that hypoxia is one of the major clinical manifestations of severe SARS-CoV-2 infection; it directly or indirectly contributes to premature neuronal aging, neuroinflammation, and neurodegeneration by altering the expression of various genes responsible for the survival of the cells. This review focuses on the interplay between COVID-19 infection, hypoxia, premature neuronal aging, and neurodegenerative diseases and provides a novel insight into the molecular mechanisms of neurodegeneration.
Collapse
Affiliation(s)
- Narmadhaa Sivagurunathan
- Molecular Pharmacology & Toxicology Laboratory, Department of Life Sciences, School of Life Sciences, Central University of Tamil Nadu, Thiruvarur - 610005, Tamil Nadu, India
| | - Latchoumycandane Calivarathan
- Molecular Pharmacology & Toxicology Laboratory, Department of Life Sciences, School of Life Sciences, Central University of Tamil Nadu, Thiruvarur - 610005, Tamil Nadu, India
| |
Collapse
|
6
|
Hajer F, Hana S, Saoussen C, Abdelhak F, Nadia B, Ameni D, Habib G, Hassen BA, Amel HK. Genetic polymorphisms in VEGFA and VEGFR2 genes associated with coronary heart disease susceptibility and severity. Mol Biol Rep 2023; 50:10169-10177. [PMID: 37924452 DOI: 10.1007/s11033-023-08899-z] [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: 07/05/2023] [Accepted: 10/04/2023] [Indexed: 11/06/2023]
Abstract
BACKGROUND Vascular endothelial growth factor A (VEGFA) is well acknowledged as a powerful angiogenesis-promoting agent mainly through its receptor VEGFR2. Ischemia stimulates VEGFA/VEGFR2 signaling pathway and elevated serum levels of VEGFA were detected in coronary heart disease (CHD) patients. The goal of the current study is to determine how four SNPs in the genes for VEGFA (rs3025039 and rs699947) and VEGFR2 (rs2305948 and rs1870377) contribute to the development of CHD. We also wanted to use the Gensini score to confirm if these four SNPs have an effect on the severity of coronary lesions. METHODS In this case-control research, we used the restriction fragment length polymorphism of the polymerase chain reaction to genotype 239 CHD patients and 200 controls. Age, sex, smoking behavior, and obesity were taken into account in the statistical analysis. RESULTS Two VEGFA/VEFGR2 signaling pathway SNPs (rs699947 and rs1870377) were found to be associated with CHD (C vs. A, P = 0.002; OR = 1.47 (1.12-1.93); A vs. T, P = 0.001; OR = 1.58 (1.17-2.13) respectively). The rs2305948 showed no allelic associations with CHD susceptibility, although we noticed a slight association under the recessive model of rs3025039 TT genotype (p = 0.023; OR = 6.41 (1.14-36.12)) only under adjusted analyses. In addition, both VEGFA SNPs (rs699947and rs3025039) were found to be associated with high Gensini score (p < 0.001). CONCLUSIONS Our research helps to shed further light on the pathophysiology of CHD. The VEGFA/VEGFR2 signaling pathway may have been downregulated, increasing CHD susceptibility and risk.
Collapse
Affiliation(s)
- Foddha Hajer
- Laboratory of Human Genome and Multifactorial Diseases (LR12ES07), Faculty of Pharmacy, University of Monastir, Monastir, Tunisia.
| | - Saoud Hana
- Laboratory of Genetics, Biodiversity and Bioresource Valorization (LR11ES41), Higher Institute of Biotechnology of Monastir, University of Monastir, Monastir, Tunisia
| | - Chouchene Saoussen
- Laboratory of Human Genome and Multifactorial Diseases (LR12ES07), Faculty of Pharmacy, University of Monastir, Monastir, Tunisia
- Department of Hematology, Fattouma Bourguiba Hospital, University of Monastir, Monastir, Tunisia
| | - Foddha Abdelhak
- Cardiology Department, Fattouma Bourguiba Hospital, University of Monastir, Monastir, Tunisia
| | - Bouzidi Nadia
- Laboratory of Human Genome and Multifactorial Diseases (LR12ES07), Faculty of Pharmacy, University of Monastir, Monastir, Tunisia
| | - Dhiflaoui Ameni
- Laboratory of Human Genome and Multifactorial Diseases (LR12ES07), Faculty of Pharmacy, University of Monastir, Monastir, Tunisia
| | - Gamra Habib
- Cardiology Department, Fattouma Bourguiba Hospital, University of Monastir, Monastir, Tunisia
| | - Ben Abdennebi Hassen
- Laboratory of Human Genome and Multifactorial Diseases (LR12ES07), Faculty of Pharmacy, University of Monastir, Monastir, Tunisia
| | - Haj Khelil Amel
- Laboratory of Human Genome and Multifactorial Diseases (LR12ES07), Faculty of Pharmacy, University of Monastir, Monastir, Tunisia
| |
Collapse
|
7
|
Pun SH, O’Neill KM, Edgar KS, Gill EK, Moez A, Naderi-Meshkin H, Malla SB, Hookham MB, Alsaggaf M, Madishetti VV, Botezatu B, King W, Brunssen C, Morawietz H, Dunne PD, Brazil DP, Medina RJ, Watson CJ, Grieve DJ. PLAC8-Mediated Activation of NOX4 Signalling Restores Angiogenic Function of Endothelial Colony-Forming Cells in Experimental Hypoxia. Cells 2023; 12:2220. [PMID: 37759443 PMCID: PMC10526321 DOI: 10.3390/cells12182220] [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: 06/19/2023] [Revised: 08/28/2023] [Accepted: 09/01/2023] [Indexed: 09/29/2023] Open
Abstract
Ischaemic cardiovascular disease is associated with tissue hypoxia as a significant determinant of angiogenic dysfunction and adverse remodelling. While cord blood-derived endothelial colony-forming cells (CB-ECFCs) hold clear therapeutic potential due to their enhanced angiogenic and proliferative capacity, their impaired functionality within the disease microenvironment represents a major barrier to clinical translation. The aim of this study was to define the specific contribution of NOX4 NADPH oxidase, which we previously reported as a key CB-ECFC regulator, to hypoxia-induced dysfunction and its potential as a therapeutic target. CB-ECFCs exposed to experimental hypoxia demonstrated downregulation of NOX4-mediated reactive oxygen species (ROS) signalling linked with a reduced tube formation, which was partially restored by NOX4 plasmid overexpression. siRNA knockdown of placenta-specific 8 (PLAC8), identified by microarray analysis as an upstream regulator of NOX4 in hypoxic versus normoxic CB-ECFCs, enhanced tube formation, NOX4 expression and hydrogen peroxide generation, and induced several key transcription factors associated with downstream Nrf2 signalling. Taken together, these findings indicated that activation of the PLAC8-NOX4 signalling axis improved CB-ECFC angiogenic functions in experimental hypoxia, highlighting this pathway as a potential target for protecting therapeutic cells against the ischaemic cardiovascular disease microenvironment.
Collapse
Affiliation(s)
- Shun Hay Pun
- Wellcome-Wolfson Institute for Experimental Medicine, Queen’s University, Belfast BT9 7AE, UK; (S.H.P.); (K.M.O.); (K.S.E.); (E.K.G.); (A.M.); (H.N.-M.); (M.B.H.); (M.A.); (V.V.M.); (B.B.); (W.K.); (D.P.B.); (R.J.M.); (C.J.W.)
| | - Karla M. O’Neill
- Wellcome-Wolfson Institute for Experimental Medicine, Queen’s University, Belfast BT9 7AE, UK; (S.H.P.); (K.M.O.); (K.S.E.); (E.K.G.); (A.M.); (H.N.-M.); (M.B.H.); (M.A.); (V.V.M.); (B.B.); (W.K.); (D.P.B.); (R.J.M.); (C.J.W.)
| | - Kevin S. Edgar
- Wellcome-Wolfson Institute for Experimental Medicine, Queen’s University, Belfast BT9 7AE, UK; (S.H.P.); (K.M.O.); (K.S.E.); (E.K.G.); (A.M.); (H.N.-M.); (M.B.H.); (M.A.); (V.V.M.); (B.B.); (W.K.); (D.P.B.); (R.J.M.); (C.J.W.)
| | - Eleanor K. Gill
- Wellcome-Wolfson Institute for Experimental Medicine, Queen’s University, Belfast BT9 7AE, UK; (S.H.P.); (K.M.O.); (K.S.E.); (E.K.G.); (A.M.); (H.N.-M.); (M.B.H.); (M.A.); (V.V.M.); (B.B.); (W.K.); (D.P.B.); (R.J.M.); (C.J.W.)
| | - Arya Moez
- Wellcome-Wolfson Institute for Experimental Medicine, Queen’s University, Belfast BT9 7AE, UK; (S.H.P.); (K.M.O.); (K.S.E.); (E.K.G.); (A.M.); (H.N.-M.); (M.B.H.); (M.A.); (V.V.M.); (B.B.); (W.K.); (D.P.B.); (R.J.M.); (C.J.W.)
| | - Hojjat Naderi-Meshkin
- Wellcome-Wolfson Institute for Experimental Medicine, Queen’s University, Belfast BT9 7AE, UK; (S.H.P.); (K.M.O.); (K.S.E.); (E.K.G.); (A.M.); (H.N.-M.); (M.B.H.); (M.A.); (V.V.M.); (B.B.); (W.K.); (D.P.B.); (R.J.M.); (C.J.W.)
| | - Sudhir B. Malla
- Patrick G Johnston Centre for Cancer Research, Queen’s University, Belfast BT9 7AE, UK; (S.B.M.); (P.D.D.)
| | - Michelle B. Hookham
- Wellcome-Wolfson Institute for Experimental Medicine, Queen’s University, Belfast BT9 7AE, UK; (S.H.P.); (K.M.O.); (K.S.E.); (E.K.G.); (A.M.); (H.N.-M.); (M.B.H.); (M.A.); (V.V.M.); (B.B.); (W.K.); (D.P.B.); (R.J.M.); (C.J.W.)
| | - Mohammed Alsaggaf
- Wellcome-Wolfson Institute for Experimental Medicine, Queen’s University, Belfast BT9 7AE, UK; (S.H.P.); (K.M.O.); (K.S.E.); (E.K.G.); (A.M.); (H.N.-M.); (M.B.H.); (M.A.); (V.V.M.); (B.B.); (W.K.); (D.P.B.); (R.J.M.); (C.J.W.)
| | - Vinuthna Vani Madishetti
- Wellcome-Wolfson Institute for Experimental Medicine, Queen’s University, Belfast BT9 7AE, UK; (S.H.P.); (K.M.O.); (K.S.E.); (E.K.G.); (A.M.); (H.N.-M.); (M.B.H.); (M.A.); (V.V.M.); (B.B.); (W.K.); (D.P.B.); (R.J.M.); (C.J.W.)
| | - Bianca Botezatu
- Wellcome-Wolfson Institute for Experimental Medicine, Queen’s University, Belfast BT9 7AE, UK; (S.H.P.); (K.M.O.); (K.S.E.); (E.K.G.); (A.M.); (H.N.-M.); (M.B.H.); (M.A.); (V.V.M.); (B.B.); (W.K.); (D.P.B.); (R.J.M.); (C.J.W.)
| | - William King
- Wellcome-Wolfson Institute for Experimental Medicine, Queen’s University, Belfast BT9 7AE, UK; (S.H.P.); (K.M.O.); (K.S.E.); (E.K.G.); (A.M.); (H.N.-M.); (M.B.H.); (M.A.); (V.V.M.); (B.B.); (W.K.); (D.P.B.); (R.J.M.); (C.J.W.)
| | - Coy Brunssen
- Division of Vascular Endothelium and Microcirculation, TUD Dresden University of Technology, 01307 Dresden, Germany; (C.B.); (H.M.)
| | - Henning Morawietz
- Division of Vascular Endothelium and Microcirculation, TUD Dresden University of Technology, 01307 Dresden, Germany; (C.B.); (H.M.)
| | - Philip D. Dunne
- Patrick G Johnston Centre for Cancer Research, Queen’s University, Belfast BT9 7AE, UK; (S.B.M.); (P.D.D.)
| | - Derek P. Brazil
- Wellcome-Wolfson Institute for Experimental Medicine, Queen’s University, Belfast BT9 7AE, UK; (S.H.P.); (K.M.O.); (K.S.E.); (E.K.G.); (A.M.); (H.N.-M.); (M.B.H.); (M.A.); (V.V.M.); (B.B.); (W.K.); (D.P.B.); (R.J.M.); (C.J.W.)
| | - Reinhold J. Medina
- Wellcome-Wolfson Institute for Experimental Medicine, Queen’s University, Belfast BT9 7AE, UK; (S.H.P.); (K.M.O.); (K.S.E.); (E.K.G.); (A.M.); (H.N.-M.); (M.B.H.); (M.A.); (V.V.M.); (B.B.); (W.K.); (D.P.B.); (R.J.M.); (C.J.W.)
| | - Chris J. Watson
- Wellcome-Wolfson Institute for Experimental Medicine, Queen’s University, Belfast BT9 7AE, UK; (S.H.P.); (K.M.O.); (K.S.E.); (E.K.G.); (A.M.); (H.N.-M.); (M.B.H.); (M.A.); (V.V.M.); (B.B.); (W.K.); (D.P.B.); (R.J.M.); (C.J.W.)
| | - David J. Grieve
- Wellcome-Wolfson Institute for Experimental Medicine, Queen’s University, Belfast BT9 7AE, UK; (S.H.P.); (K.M.O.); (K.S.E.); (E.K.G.); (A.M.); (H.N.-M.); (M.B.H.); (M.A.); (V.V.M.); (B.B.); (W.K.); (D.P.B.); (R.J.M.); (C.J.W.)
| |
Collapse
|
8
|
Oliveira RHM, Annex BH, Popel AS. Endothelial cells signaling and patterning under hypoxia: a mechanistic integrative computational model including the Notch-Dll4 pathway. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.05.03.539270. [PMID: 37205581 PMCID: PMC10187169 DOI: 10.1101/2023.05.03.539270] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/21/2023]
Abstract
Several signaling pathways are activated during hypoxia to promote angiogenesis, leading to endothelial cell patterning, interaction, and downstream signaling. Understanding the mechanistic signaling differences between normoxia and hypoxia can guide therapies to modulate angiogenesis. We present a novel mechanistic model of interacting endothelial cells, including the main pathways involved in angiogenesis. We calibrate and fit the model parameters based on well-established modeling techniques. Our results indicate that the main pathways involved in the patterning of tip and stalk endothelial cells under hypoxia differ, and the time under hypoxia affects how a reaction affects patterning. Interestingly, the interaction of receptors with Neuropilin1 is also relevant for cell patterning. Our simulations under different oxygen concentrations indicate time- and oxygen-availability-dependent responses for the two cells. Following simulations with various stimuli, our model suggests that factors such as period under hypoxia and oxygen availability must be considered for pattern control. This project provides insights into the signaling and patterning of endothelial cells under hypoxia, contributing to studies in the field.
Collapse
Affiliation(s)
- Rebeca Hannah M Oliveira
- Department of Biomedical Engineering, School of Medicine, Johns Hopkins University, Baltimore, Maryland, 21205, USA
| | - Brian H Annex
- Medical College of Georgia, Augusta University, Augusta, Georgia, USA
| | - Aleksander S Popel
- Department of Biomedical Engineering, School of Medicine, Johns Hopkins University, Baltimore, Maryland, 21205, USA
| |
Collapse
|
9
|
Circulating Biomarkers in Patients With Locally Advanced or Metastatic Renal Cell Carcinoma Treated With Everolimus in the Pre-nephrectomy Setting. Clin Oncol (R Coll Radiol) 2023; 35:e245-e255. [PMID: 36526521 DOI: 10.1016/j.clon.2022.11.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Revised: 09/28/2022] [Accepted: 11/18/2022] [Indexed: 12/23/2022]
Abstract
Many drugs are available in renal cell carcinoma (RCC), yet clinicians are still looking for predictive biomarkers of disease recurrence or progression supporting more personalised treatments. An assessment of circulating biomarkers over time was carried out in this French, open-label, single-arm, multicentre trial conducted in 25 patients with either locally advanced (n = 14) or metastatic RCC (n = 11) who received everolimus (10 mg daily) for 6 weeks prior to nephrectomy (NEORAD, NCT01715935). Circulating biomarkers, including circulating tumour cells, haematopoietic and endothelial cells, plasma angiogenesis and inflammatory markers were quantified at baseline, upon everolimus and post-nephrectomy. We assessed tumour burden, objective response rate upon RECIST1.1, disease-free survival (DFS) and progression-free survival (PFS). The correlation between circulating biomarkers was evaluated with multiple factor analysis and biomarker association with DFS/PFS by Cox regression. No objective response rate was obtained before nephrectomy. Upon everolimus, neutrophils, platelets and sVEGFR2 significantly decreased. We did not find any association between circulating biomarkers and DFS/PFS, but patients with the highest tumour burden at baseline had significantly higher plasma levels of interleukin-6, an inflammatory circulating biomarker, and lower levels of sVEGFR2, related to angiogenesis. Further understanding of the link between these circulating biomarkers could help to optimise drug combinations in RCC.
Collapse
|
10
|
Chang J, Sun Y, Meng X, Zeng F, Wang X. EGFL7 affects the migration of epidermal stem cells in refractory diabetic wounds by regulating Notch signaling pathway. Regen Med 2023; 18:137-153. [PMID: 36530156 DOI: 10.2217/rme-2022-0123] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Aim: This study aimed to explore the role of EGFL7 in the healing process of refractory diabetic wounds. Methods: Epidermal stem cells (ESCs) were isolated from healthy mice and diabetic mice, identified by immunofluorescence, transfected with EGFL7 overexpression and silencing lentiviral vectors, and treated with Notch pathway inhibitor (DAPT). Results: SiEGFL7 significantly inhibited the proliferation, invasion and migration of ESCs of healthy mice. DAPT prominently inhibited the expressions of Notch1, Notch2, Hes1 and Jag1 in ESCs of healthy mice induced by overexpressed EGFL7. Overexpressed EGFL7 promoted wound healing in diabetic mice with refractory wounds. Conclusion: EGFL7 affects the proliferation and migration of ESCs in refractory diabetic wounds by regulating the Notch signaling pathway.
Collapse
Affiliation(s)
- Jinyuan Chang
- Department of Burn and Plastic Surgery, The Second Xiangya Hospital of Central South University, No. 139, Shaoshan South Road, Furong District, Changsha, Hunan, 410011, China
| | - Yang Sun
- Department of Burn and Plastic Surgery, The Second Xiangya Hospital of Central South University, No. 139, Shaoshan South Road, Furong District, Changsha, Hunan, 410011, China
| | - Xianxi Meng
- Department of Burn and Plastic Surgery, The Second Xiangya Hospital of Central South University, No. 139, Shaoshan South Road, Furong District, Changsha, Hunan, 410011, China
| | - Fanglin Zeng
- Department of Burn and Plastic Surgery, The Second Xiangya Hospital of Central South University, No. 139, Shaoshan South Road, Furong District, Changsha, Hunan, 410011, China
| | - Xiancheng Wang
- Department of Burn and Plastic Surgery, The Second Xiangya Hospital of Central South University, No. 139, Shaoshan South Road, Furong District, Changsha, Hunan, 410011, China
| |
Collapse
|
11
|
Abel T, Moodley J, Khaliq OP, Naicker T. Vascular Endothelial Growth Factor Receptor 2: Molecular Mechanism and Therapeutic Potential in Preeclampsia Comorbidity with Human Immunodeficiency Virus and Severe Acute Respiratory Syndrome Coronavirus 2 Infections. Int J Mol Sci 2022; 23:ijms232213752. [PMID: 36430232 PMCID: PMC9691176 DOI: 10.3390/ijms232213752] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Revised: 11/05/2022] [Accepted: 11/07/2022] [Indexed: 11/11/2022] Open
Abstract
This review explored the role of vascular endothelial growth factor receptor-2 (VEGFR-2) in the synergy of preeclampsia (PE), human immunodeficiency virus (HIV), and severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infections. Downregulation of VEGFR-2 in PE promotes endothelial dysfunction and prevents endothelial cell (EC) migration, proliferation, and differentiation. The HIV-1 accessory protein, tat (trans-activator of transcription), prevents VEGFR-2 signaling via the vascular endothelial growth factor A (VEGF-A) ligand. Combined antiretroviral therapy (cART) may cause immune reconstitution, impaired decidualization, and endothelial injury, thus may be a risk factor for PE development. The VEGF/VEGFR-2 interaction may be associated with SARS-CoV-2-related pulmonary oedema. Endothelial dysfunction and heightened inflammation are both associated with PE, HIV, and SARS-CoV-2 infection; therefore, it is plausible that both characteristics may be exacerbated in the synergy of these events. In addition, this review explored microRNAs (miR) regulating VEGFR-2. An overexpression of miR-126 is evident in PE, HIV, and SARS-CoV-2 infection; thus, modulating the expression of miR-126 may be a therapeutic strategy. However, the involvement of microRNAs in PE, HIV, and SARS-CoV-2 infection needs further investigating. Since these conditions have been evaluated independently, this review attempts to predict their clinical manifestations in their synergy, as well as independently; thereby providing a platform for early diagnosis and therapeutic potential in PE, HIV, and SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Tashlen Abel
- Women’s Health and HIV Research Group, Department of Obstetrics & Gynaecology, School of Clinical Medicine, College of Health Sciences, University of KwaZulu-Natal, Durban 4000, South Africa
| | - Jagidesa Moodley
- Women’s Health and HIV Research Group, Department of Obstetrics & Gynaecology, School of Clinical Medicine, College of Health Sciences, University of KwaZulu-Natal, Durban 4000, South Africa
| | - Olive P. Khaliq
- Department of Paediatrics and Child Health, Faculty of Health Sciences, The University of the Free State, Bloemfontein 9300, South Africa
| | - Thajasvarie Naicker
- Optics and Imaging Centre, Doris Duke Medical Research Institution, College of Health Sciences, University of KwaZulu-Natal, Durban 4000, South Africa
- Correspondence:
| |
Collapse
|
12
|
Phillippi JA. On vasa vasorum: A history of advances in understanding the vessels of vessels. SCIENCE ADVANCES 2022; 8:eabl6364. [PMID: 35442731 PMCID: PMC9020663 DOI: 10.1126/sciadv.abl6364] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Accepted: 03/01/2022] [Indexed: 05/09/2023]
Abstract
The vasa vasorum are a vital microvascular network supporting the outer wall of larger blood vessels. Although these dynamic microvessels have been studied for centuries, the importance and impact of their functions in vascular health and disease are not yet fully realized. There is now rich knowledge regarding what local progenitor cell populations comprise and cohabitate with the vasa vasorum and how they might contribute to physiological and pathological changes in the network or its expansion via angiogenesis or vasculogenesis. Evidence of whether vasa vasorum remodeling incites or governs disease progression or is a consequence of cardiovascular pathologies remains limited. Recent advances in vasa vasorum imaging for understanding cardiovascular disease severity and pathophysiology open the door for theranostic opportunities. Approaches that strive to control angiogenesis and vasculogenesis potentiate mitigation of vasa vasorum-mediated contributions to cardiovascular diseases and emerging diseases involving the microcirculation.
Collapse
Affiliation(s)
- Julie A. Phillippi
- Department of Cardiothoracic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Department of Bioengineering, University of Pittsburgh Swanson School of Engineering, Pittsburgh, PA, USA
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| |
Collapse
|
13
|
Klomjit N, Zhu XY, Massat AE, Pawar AS, Conley S, Puranik AS, Ferguson CM, Kim SR, Tang H, Jordan K, Saadiq I, Lerman A, Grande JP, Textor SC, Lerman LO. Microvascular remodeling and altered angiogenic signaling in human kidneys distal to occlusive atherosclerotic renal artery stenosis. Nephrol Dial Transplant 2022; 37:1844-1856. [PMID: 35451482 PMCID: PMC9494086 DOI: 10.1093/ndt/gfac156] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND Renal artery stenosis (RAS) is an important cause of chronic kidney disease and secondary hypertension. In animal models, renal ischemia leads to downregulation of growth-factor expression and loss of intrarenal microcirculation. However, little is known about the sequelae of large vessel occlusive disease on the microcirculation within human kidneys. METHOD This study included 5 patients who underwent nephrectomy due to renovascular occlusion, and 7 non-stenotic discarded donor kidneys (4 deceased donors). Micro-computed tomography was performed to assess microvascular spatial densities and tortuosity, an index of microvascular immaturity. Renal protein expression, gene expression, and histology were studied in-vitro using immunoblotting, polymerase-chain-reaction, and staining. RESULTS RAS demonstrated loss of medium-sized vessels (0.2-0.3mm) compared to donor kidneys (p = 0.037) and increased microvascular tortuosity. RAS kidneys had greater protein expression of angiopoietin-1, hypoxia-inducible factor (HIF)-1α, and thrombospondin (TSP)-1, but lower protein expression of vascular endothelial growth factor (VEGF) than donor kidneys. Renal fibrosis, loss of peritubular capillaries (PTC) and pericyte detachment were greater in RAS, yet they had more newly-formed PTC than donor kidneys. Therefore, our study quantified significant microvascular remodeling in the post-stenotic human kidney. RAS induced renal microvascular loss, vascular remodeling, and fibrosis. Despite downregulated VEGF, stenotic kidneys upregulated compensatory angiogenic pathways related to angiopoietin-1. CONCLUSIONS These observations underscore the nature of human RAS as a microvascular disease distal to main vessel stenosis, and support therapeutic strategies directly targeting the post-stenotic kidney microcirculation in patients with RAS.
Collapse
Affiliation(s)
- Nattawat Klomjit
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, MN, USA.,Division of Nephrology and Hypertension, University of Minnesota, Minneapolis, MN, USA
| | - Xiang-Yang Zhu
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, MN, USA
| | | | - Aditya S Pawar
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, MN, USA
| | - Sabena Conley
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, MN, USA
| | - Amrutesh S Puranik
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, MN, USA
| | | | - Seo Rin Kim
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, MN, USA
| | - Hui Tang
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, MN, USA
| | - Kyra Jordan
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, MN, USA
| | - Ishran Saadiq
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, MN, USA
| | - Amir Lerman
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN, USA
| | - Joseph P Grande
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, USA
| | - Stephen C Textor
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, MN, USA
| | - Lilach O Lerman
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, MN, USA
| |
Collapse
|
14
|
Adam RJ, Williams AC, Kriegel AJ. Comparison of the Surgical Resection and Infarct 5/6 Nephrectomy Rat Models of Chronic Kidney Disease. Am J Physiol Renal Physiol 2022; 322:F639-F654. [PMID: 35379002 DOI: 10.1152/ajprenal.00398.2021] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
The 5/6 nephrectomy rat remnant kidney model is commonly employed to study chronic kidney disease (CKD). This model requires removal of one whole kidney and two-thirds of the other. The two most common ways of producing the remnant kidney are surgical resection of poles, known as the polectomy (Pol) model, or ligation of upper and lower renal arterial branches, resulting in pole infarction (Inf). These models have much in common, but also major phenotypic differences, and thus respectively model unique aspects of human CKD. The purpose of this review is to summarize phenotypic similarities and differences between these two models and their relation to human CKD, while emphasizing their vascular phenotype. In this article we review studies that have evaluated arterial blood pressure, the renin-angiotensin-aldosterone-system (RAAS), autoregulation, nitric oxide, single nephron physiology, angiogenic and anti-angiogenic factors, and capillary rarefaction in these two models. Phenotypic similarities: both models spontaneously develop hallmarks of human CKD including uremia, fibrosis, capillary rarefaction, and progressive renal function decline. They both undergo whole-organ hypertrophy, hyperfiltration of functional nephrons, reduced renal expression of angiogenic factor VEGF, increased renal expression of the anti-angiogenic thrombospondin-1, impaired renal autoregulation, and abnormal vascular nitric oxide physiology. Key phenotypic differences: the Inf model develops rapid-onset, moderate-to-severe systemic hypertension, and the Pol model early normotension followed by mild-to-moderate hypertension. The Inf rat has a markedly more active renin-angiotensin-aldosterone-system. Comparison of these two models facilitates understanding of how they can be utilized for studying CKD pathophysiology (e.g., RAAS dependent or independent pathology).
Collapse
Affiliation(s)
- Ryan J Adam
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI, United States.,Cardiovascular Center, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Adaysha C Williams
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Alison J Kriegel
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI, United States.,Department of Pediatrics, Medical College of Wisconsin, Milwaukee, WI, United States.,Cardiovascular Center, Medical College of Wisconsin, Milwaukee, WI, United States.,Center of Systems Molecular Medicine, Medical College of Wisconsin, Milwaukee, WI, United States
| |
Collapse
|
15
|
Pro-Calcific Environment Impairs Ischaemia-Driven Angiogenesis. Int J Mol Sci 2022; 23:ijms23063363. [PMID: 35328786 PMCID: PMC8954938 DOI: 10.3390/ijms23063363] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Revised: 03/14/2022] [Accepted: 03/17/2022] [Indexed: 02/04/2023] Open
Abstract
Peripheral arterial disease (PAD) is characterised by accelerated arterial calcification and impairment in angiogenesis. Studies implicate vascular calcification as a contributor to PAD, but the mechanisms remain unclear. We aimed to determine the effect of calcification on ischaemia-driven angiogenesis. Human coronary artery endothelial cells (ECs) were treated with calcification medium (CM: CaCl2 2.7 mM, Na2PO4 2.0 mM) for 24 h and exposed to normoxia (5% CO2) or hypoxia (1.2% O2; 5% CO2 balanced with N2). In normoxia, CM significantly inhibited tubule formation and migration and upregulated calcification markers of ALP, BMP2, and Runx2. CM elevated levels of calcification-protective gene OPG, demonstrating a compensatory mechanism by ECs. CM failed to induce pro-angiogenic regulators VEGFA and HIF-1α in hypoxia and further suppressed the phosphorylation of endothelial nitric oxide synthase (eNOS) that is essential for vascular function. In vivo, osteoprotegerin-deficient mice (OPG−/−), a calcification model, were subjected to hind-limb ischaemia (HLI) surgery. OPG−/− mice displayed elevated serum alkaline phosphatase (ALP) activity compared to wild-type controls. OPG−/− mice experienced striking reductions in blood-flow reperfusion in both 8-week-old and 6-month-old mice post-HLI. This coincided with significant impairment in tissue ischaemia and reduced limb function as assessed by clinical scoring (Tarlov). This study demonstrated for the first time that a pro-calcific environment is detrimental to ischaemia-driven angiogenesis. The degree of calcification in patients with PAD can often be a limiting factor with the use of standard therapies. These highly novel findings require further studies for full elucidation of the mechanisms involved and have implications for the development of therapies to suppress calcification in PAD.
Collapse
|
16
|
Emergent players in renovascular disease. Clin Sci (Lond) 2022; 136:239-256. [PMID: 35129198 DOI: 10.1042/cs20210509] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Revised: 01/25/2022] [Accepted: 01/26/2022] [Indexed: 02/07/2023]
Abstract
Renovascular disease (RVD) remains a common etiology of secondary hypertension. Recent clinical trials revealed unsatisfactory therapeutic outcomes of renal revascularization, leading to extensive investigation to unravel key pathophysiological mechanisms underlying irreversible functional loss and structural damage in the chronically ischemic kidney. Research studies identified complex interactions among various players, including inflammation, fibrosis, mitochondrial injury, cellular senescence, and microvascular remodeling. This interplay resulted in a shift of our understanding of RVD from a mere hemodynamic disorder to a pro-inflammatory and pro-fibrotic pathology strongly influenced by systemic diseases like metabolic syndrome (MetS), hypertension, diabetes mellitus, and hyperlipidemia. Novel diagnostic approaches have been tested for early detection and follow-up of RVD progression, using new imaging techniques and biochemical markers of renal injury and dysfunction. Therapies targeting some of the pathological pathways governing the development of RVD have shown promising results in animal models, and a few have moved from bench to clinical research. This review summarizes evolving understanding in chronic ischemic kidney injury.
Collapse
|
17
|
Waller JP, Howell JA, Peterson H, George EM, Bidwell GL. Elastin-Like Polypeptide: VEGF-B Fusion Protein for Treatment of Preeclampsia. Hypertension 2021; 78:1888-1901. [PMID: 34719237 PMCID: PMC8585700 DOI: 10.1161/hypertensionaha.121.17713] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2021] [Accepted: 09/30/2021] [Indexed: 11/16/2022]
Abstract
[Figure: see text].
Collapse
Affiliation(s)
- Jamarius P. Waller
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, 2500 North State Street, Jackson, MS. 39216
| | - John Aaron Howell
- Department of Neurology, University of Mississippi Medical Center, 2500 North State Street, Jackson, MS. 39216
| | - Hali Peterson
- Department of Medicine, University of Mississippi Medical Center, 2500 North State Street, Jackson, MS. 39216
| | - Eric M. George
- Department of Physiology, University of Mississippi Medical Center, 2500 North State Street, Jackson, MS. 39216
- Department of Cell and Molecular Biology, University of Mississippi Medical Center, 2500 North State Street, Jackson, MS. 39216
| | - Gene L. Bidwell
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, 2500 North State Street, Jackson, MS. 39216
- Department of Neurology, University of Mississippi Medical Center, 2500 North State Street, Jackson, MS. 39216
- Department of Cell and Molecular Biology, University of Mississippi Medical Center, 2500 North State Street, Jackson, MS. 39216
| |
Collapse
|
18
|
Janaszak-Jasiecka A, Siekierzycka A, Płoska A, Dobrucki IT, Kalinowski L. Endothelial Dysfunction Driven by Hypoxia-The Influence of Oxygen Deficiency on NO Bioavailability. Biomolecules 2021; 11:biom11070982. [PMID: 34356605 PMCID: PMC8301841 DOI: 10.3390/biom11070982] [Citation(s) in RCA: 53] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 06/25/2021] [Accepted: 07/02/2021] [Indexed: 12/12/2022] Open
Abstract
Cardiovascular diseases (CVDs) are the leading cause of death worldwide. The initial stage of CVDs is characterized by endothelial dysfunction, defined as the limited bioavailability of nitric oxide (NO). Thus, any factors that interfere with the synthesis or metabolism of NO in endothelial cells are involved in CVD pathogenesis. It is well established that hypoxia is both the triggering factor as well as the accompanying factor in cardiovascular disease, and diminished tissue oxygen levels have been reported to influence endothelial NO bioavailability. In endothelial cells, NO is produced by endothelial nitric oxide synthase (eNOS) from L-Arg, with tetrahydrobiopterin (BH4) as an essential cofactor. Here, we discuss the mechanisms by which hypoxia affects NO bioavailability, including regulation of eNOS expression and activity. What is particularly important is the fact that hypoxia contributes to the depletion of cofactor BH4 and deficiency of substrate L-Arg, and thus elicits eNOS uncoupling-a state in which the enzyme produces superoxide instead of NO. eNOS uncoupling and the resulting oxidative stress is the major driver of endothelial dysfunction and atherogenesis. Moreover, hypoxia induces impairment in mitochondrial respiration and endothelial cell activation; thus, oxidative stress and inflammation, along with the hypoxic response, contribute to the development of endothelial dysfunction.
Collapse
Affiliation(s)
- Anna Janaszak-Jasiecka
- Department of Medical Laboratory Diagnostics—Fahrenheit Biobank BBMRI.pl, Medical University of Gdansk, 80-211 Gdansk, Poland; (A.J.-J.); (A.S.); (A.P.)
- Biobanking and Biomolecular Resources Research Infrastructure Poland (BBMRI.pl), 80-211 Gdansk, Poland
| | - Anna Siekierzycka
- Department of Medical Laboratory Diagnostics—Fahrenheit Biobank BBMRI.pl, Medical University of Gdansk, 80-211 Gdansk, Poland; (A.J.-J.); (A.S.); (A.P.)
- Laboratory of Trace Elements Neurobiology, Institute of Pharmacology, Polish Academy of Sciences, 31-343 Krakow, Poland
| | - Agata Płoska
- Department of Medical Laboratory Diagnostics—Fahrenheit Biobank BBMRI.pl, Medical University of Gdansk, 80-211 Gdansk, Poland; (A.J.-J.); (A.S.); (A.P.)
- Biobanking and Biomolecular Resources Research Infrastructure Poland (BBMRI.pl), 80-211 Gdansk, Poland
| | - Iwona T. Dobrucki
- University of Illinois at Urbana-Champaign Beckman Institute for Advanced Science and Technology, 405 N Mathews Ave, MC-251, Urbana, IL 61801, USA;
- Department of Bioengineering, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - Leszek Kalinowski
- Department of Medical Laboratory Diagnostics—Fahrenheit Biobank BBMRI.pl, Medical University of Gdansk, 80-211 Gdansk, Poland; (A.J.-J.); (A.S.); (A.P.)
- Biobanking and Biomolecular Resources Research Infrastructure Poland (BBMRI.pl), 80-211 Gdansk, Poland
- BioTechMed Centre, Department of Mechanics of Materials and Structures, Gdansk University of Technology, 80-233 Gdansk, Poland
- Correspondence:
| |
Collapse
|
19
|
Lee AR, Baek SM, Lee SW, Kim TU, Han JE, Bae S, Park SJ, Kim TH, Jeong KS, Choi SK, Park JK. Nuclear VEGFR-2 Expression of Hepatocytes Is Involved in Hepatocyte Proliferation and Liver Regeneration During Chronic Liver Injury. In Vivo 2021; 35:1473-1483. [PMID: 33910825 DOI: 10.21873/invivo.12400] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Revised: 03/06/2021] [Accepted: 03/12/2021] [Indexed: 11/10/2022]
Abstract
BACKGROUND/AIM The pathological role of vascular endothelial growth factor receptor 2 (VEGFR-2) in chronic liver injury and liver regeneration is not fully understood. This study analysed the role of VEGFR-2 in liver fibrosis and its regeneration process. MATERIALS AND METHODS We administered intraperitoneally 50 mg/kg to 300 mg/kg thioacetamide (TAA) to 9-week-old male mice for 17 weeks. We measured levels of VEGFR-2 protein and identified the location of cells that specifically express VEGFR-2. RESULTS VEGFR-2 is rarely expressed in normal hepatocytes. However, high VEGFR-2 expression in liver sinusoidal endothelial cells was noted in the TAA group. Conversely, the group that experienced regeneration from liver fibrosis showed significantly higher VEGFR-2 expression in the nucleus of hepatocytes compared to the other groups. CONCLUSION VEGFR-2 plays a pivotal role in the nucleus of hepatocytes during liver regeneration and VEGFR-2 may be closely related to cell division. Therefore, VEGFR-2 may be a new therapeutic target for liver regeneration.
Collapse
Affiliation(s)
- A-Rang Lee
- Department of Veterinary Pathology, College of Veterinary Medicine, Kyungpook National University, Daegu, Republic of Korea
| | - Su-Min Baek
- Department of Veterinary Pathology, College of Veterinary Medicine, Kyungpook National University, Daegu, Republic of Korea
| | - Seoung-Woo Lee
- Department of Veterinary Pathology, College of Veterinary Medicine, Kyungpook National University, Daegu, Republic of Korea
| | - Tae-Un Kim
- Department of Veterinary Pathology, College of Veterinary Medicine, Kyungpook National University, Daegu, Republic of Korea
| | - Jee Eun Han
- College of Veterinary Medicine, Kyungpook National University, Daegu, Republic of Korea
| | - Seulgi Bae
- College of Veterinary Medicine, Kyungpook National University, Daegu, Republic of Korea
| | - Sang-Joon Park
- College of Veterinary Medicine, Kyungpook National University, Daegu, Republic of Korea
| | - Tae-Hwan Kim
- Department of Veterinary Pathology, College of Veterinary Medicine, Kyungpook National University, Daegu, Republic of Korea
| | - Kyu-Shik Jeong
- Department of Veterinary Pathology, College of Veterinary Medicine, Kyungpook National University, Daegu, Republic of Korea.,Stem Cell Therapeutic Research Institute, Kyungpook National University, Daegu, Republic of Korea
| | - Seong-Kyoon Choi
- Core Protein Resources Center, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu, Republic of Korea
| | - Jin-Kyu Park
- Department of Veterinary Pathology, College of Veterinary Medicine, Kyungpook National University, Daegu, Republic of Korea; .,Stem Cell Therapeutic Research Institute, Kyungpook National University, Daegu, Republic of Korea
| |
Collapse
|
20
|
Inflammation and Oxidative Damage in Ischaemic Renal Disease. Antioxidants (Basel) 2021; 10:antiox10060845. [PMID: 34070611 PMCID: PMC8227971 DOI: 10.3390/antiox10060845] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2021] [Revised: 05/12/2021] [Accepted: 05/19/2021] [Indexed: 12/15/2022] Open
Abstract
Ischaemic renal disease as result of atherosclerotic renovascular disease activates a complex biological response that ultimately leads to fibrosis and chronic kidney disease. Large randomised control trials have shown that renal revascularisation in patients with atherosclerotic renal artery disease does not confer any additional benefit to medical therapy alone. This is likely related to the activation of complex pathways of oxidative stress, inflammatory cytokines and fibrosis due to atherosclerotic disease and hypoxic injury due to reduced renal blood flow. New evidence from pre-clinical trials now indicates a role for specific targeted therapeutic interventions to counteract this complex pathogenesis. This evidence now suggests that the focus for those with atherosclerotic renovascular disease should be a combination of revascularisation and renoprotective therapies that target the renal tissue response to ischaemia, reduce the inflammatory infiltrate and prevent or reduce the fibrosis.
Collapse
|
21
|
A dose-escalating toxicology study of the candidate biologic ELP-VEGF. Sci Rep 2021; 11:6216. [PMID: 33737643 PMCID: PMC7973730 DOI: 10.1038/s41598-021-85693-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Accepted: 03/04/2021] [Indexed: 01/31/2023] Open
Abstract
Vascular Endothelial Growth Factor (VEGF), a key mediator of angiogenesis and vascular repair, is reduced in chronic ischemic renal diseases, leading to microvascular rarefaction and deterioration of renal function. We developed a chimeric fusion of human VEGF-A121 with the carrier protein Elastin-like Polypeptide (ELP-VEGF) to induce therapeutic angiogenesis via targeted renal VEGF therapy. We previously showed that ELP-VEGF improves renal vascular density, renal fibrosis, and renal function in swine models of chronic renal diseases. However, VEGF is a potent cytokine that induces angiogenesis and increases vascular permeability, which could cause undesired off-target effects or be deleterious in a patient with a solid tumor. Therefore, the current study aims to define the toxicological profile of ELP-VEGF and assess its risk for exacerbating tumor progression and vascularity using rodent models. A dose escalating toxicology assessment of ELP-VEGF was performed by administering a bolus intravenous injection at doses ranging from 0.1 to 200 mg/kg in Sprague Dawley (SD) rats. Blood pressure, body weight, and glomerular filtration rate (GFR) were quantified longitudinally, and terminal blood sampling and renal vascular density measurements were made 14 days after treatment. Additionally, the effects of a single administration of ELP-VEGF (0.1-10 mg/kg) on tumor growth rate, mass, and vascular density were examined in a mouse model of breast cancer. At doses up to 200 mg/kg, ELP-VEGF had no effect on body weight, caused no changes in plasma or urinary markers of renal injury, and did not induce renal fibrosis or other histopathological findings in SD rats. At the highest doses (100-200 mg/kg), ELP-VEGF caused an acute, transient hypotension (30 min), increased GFR, and reduced renal microvascular density 14 days after injection. In a mouse tumor model, ELP-VEGF did not affect tumor growth rate or tumor mass, but analysis of tumor vascular density by micro-computed tomography (μCT) revealed significant, dose dependent increases in tumor vascularity after ELP-VEGF administration. ELP-VEGF did not induce toxicity in the therapeutic dosing range, and doses one hundred times higher than the expected maximum therapeutic dose were needed to observe any adverse signs in rats. In breast tumor-bearing mice, ELP-VEGF therapy induced a dose-dependent increase in tumor vascularity, demanding caution for potential use in a patient suffering from kidney disease but with known or suspected malignancy.
Collapse
|
22
|
Jin M, Lu J, Fei X, Lu Z, Quan K, Liu Y, Chu M, Di R, Wei C, Wang H. Selection Signatures Analysis Reveals Genes Associated with High-Altitude Adaptation in Tibetan Goats from Nagqu, Tibet. Animals (Basel) 2020; 10:ani10091599. [PMID: 32911823 PMCID: PMC7552128 DOI: 10.3390/ani10091599] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2020] [Revised: 08/28/2020] [Accepted: 09/04/2020] [Indexed: 12/18/2022] Open
Abstract
Simple Summary In the process of domestication, goats have undergone long-term artificial and natural selection, leading to differences among goat breeds and leaving different selection traces on the genome. However, the genetic components underlying high-altitude adaptation remain largely unknown. Here, we genotyped four goat breeds using the Illumina Caprine 50K single nucleotide polymorphism (SNP) Chip. One highland breed (Tibetan goat) compared with three lowland breeds (Huanghuai goat, Taihang goat and Xinjiang goat) to identify the molecular basis of high-altitude adaptation. So, we investigated selection signatures using the di statistic of four goat breeds and some genes in Tibetan goats related to high-altitude adaptation were identified. In addition, q-PCR validated the gene expression level in Tibetan goats and Huanghuai goats. This information may be valuable for the study of the genetic uniqueness of Tibetan goats and increased understanding of the hypoxic adaptation mechanism of Tibetan goats on the plateau. Abstract Tibetan goat is an ancient breed, which inhabits the adverse conditions of the plateaus in China. To investigate the role of selection in shaping its genomes, we genotyped Tibetan goats (Nagqu Prefecture, above 4500 m) and three lowland populations (Xinjiang goats, Taihang goats and Huanghuai goats). The result of PCA, neighbor-joining (N-J) tree and model-based clustering showed that the genetic structure between the Tibetan goat and the three lowland populations has significant difference. As demonstrated by the di statistic, we found that some genes were related to the high-altitude adaptation of Tibetan goats. Functional analysis revealed that these genes were enriched in the VEGF (vascular endothelial growth factor) signaling pathway and melanoma, suggesting that nine genes (FGF2, EGFR, AKT1, PTEN, MITF, ENPEP, SIRT6, KDR, and CDC42) might have important roles in the high-altitude adaptation of Nagqu Tibetan goats. We also found that the LEPR gene was under the strongest selection (di value = 16.70), and it could induce upregulation of the hypoxic ventilatory response. In addition, five genes (LEPR, LDB1, EGFR, NOX4 and FGF2) with high di values were analyzed using q-PCR. Among them, we found that LEPR, LDB1 and FGF2 exhibited higher expression in the lungs of the Tibetan goats; LEPR, EGFR and LDB1 exhibited higher expression in the hearts of the Huanghuai goat. Our results suggest that LEPR, LDB1, EGFR and FGF2 genes may be related to the high-altitude adaptation of the goats. These findings improve our understanding of the selection of the high-altitude adaptability of the Nagqu Tibetan goats and provide new theoretical knowledge for the conservation and utilization of germplasm resources.
Collapse
Affiliation(s)
- Meilin Jin
- Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing 100193, China; (M.J.); (X.F.); (M.C.); (R.D.)
| | - Jian Lu
- National Animal Husbandry Service, Beijing 100193, China;
| | - Xiaojuan Fei
- Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing 100193, China; (M.J.); (X.F.); (M.C.); (R.D.)
| | - Zengkui Lu
- Lanzhou Institute of Husbandry and Pharmaceutical Sciences, Chinese Academy of Agricultural Sciences, Lanzhou 730050, China;
| | - Kai Quan
- College of Animal Science and Technology, Henan University of Animal Husbandry and Economy, Zhengzhou 450046, China;
| | - Yongbin Liu
- Inner Mongolia Academy of Animal Husbandry Science, Hohhot 010031, China;
| | - Mingxing Chu
- Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing 100193, China; (M.J.); (X.F.); (M.C.); (R.D.)
| | - Ran Di
- Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing 100193, China; (M.J.); (X.F.); (M.C.); (R.D.)
| | - Caihong Wei
- Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing 100193, China; (M.J.); (X.F.); (M.C.); (R.D.)
- Correspondence: (C.W.); (H.W.)
| | - Huihua Wang
- Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing 100193, China; (M.J.); (X.F.); (M.C.); (R.D.)
- Correspondence: (C.W.); (H.W.)
| |
Collapse
|
23
|
Yuan X, Lu H, Zhao A, Ding Y, Min Q, Wang R. Transcriptional regulation of CYP3A4 by nuclear receptors in human hepatocytes under hypoxia. Drug Metab Rev 2020; 52:225-234. [PMID: 32270716 DOI: 10.1080/03602532.2020.1733004] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
The human hepatic cytochrome P-450 3A4 (CYP3A4), recognized as a multifunctional enzyme, has a wide range of substrates including commonly used drugs. Previous investigations demonstrated that the expression of CYP3A4 in human hepatocytes could be regulated by some nuclear receptors (NRs) at transcriptional level under diverse situations. The significance of oxygen on CYP3A4-mediated metabolism seems notable while the regulatory mode of CYP3A4 in the particular case still remains elusive. Recently, striking evidence has emerged that both CYP3A4 and its regulator NR could be inhibited by exposure to hypoxia. Therefore, it is of great importance to elucidate whether and how these NRs act in the transcriptional regulation of CYP3A4 in human hepatocytes under hypoxic conditions. In this review, we mainly summarized transcriptional regulation of the pivotal enzyme CYP3A4 by NRs and explored the possible regulatory pathways of CYP3A4 via these major NRs under hypoxia, expecting to provide favorable evidence for further clinical guidance under such pathological situations.
Collapse
Affiliation(s)
- Xuechun Yuan
- Key Laboratory of the Plateau Environmental Damage Control, The 940th Hospital of Joint Logistics Support Force of Chinese People's Liberation Army, Lanzhou, China.,College of Pharmacy, Lanzhou University, Lanzhou, China
| | - Hui Lu
- Key Laboratory of the Plateau Environmental Damage Control, The 940th Hospital of Joint Logistics Support Force of Chinese People's Liberation Army, Lanzhou, China
| | - Anpeng Zhao
- Key Laboratory of the Plateau Environmental Damage Control, The 940th Hospital of Joint Logistics Support Force of Chinese People's Liberation Army, Lanzhou, China
| | - Yidan Ding
- Key Laboratory of the Plateau Environmental Damage Control, The 940th Hospital of Joint Logistics Support Force of Chinese People's Liberation Army, Lanzhou, China.,College of Pharmacy, Lanzhou University, Lanzhou, China
| | - Qiong Min
- Pharmacy department, Gansu Provincial Cancer Hospital, Lanzhou, China
| | - Rong Wang
- Key Laboratory of the Plateau Environmental Damage Control, The 940th Hospital of Joint Logistics Support Force of Chinese People's Liberation Army, Lanzhou, China.,College of Pharmacy, Lanzhou University, Lanzhou, China
| |
Collapse
|
24
|
Zhou C, Zou QY, Jiang YZ, Zheng J. Role of oxygen in fetoplacental endothelial responses: hypoxia, physiological normoxia, or hyperoxia? Am J Physiol Cell Physiol 2020; 318:C943-C953. [PMID: 32267717 DOI: 10.1152/ajpcell.00528.2019] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
During pregnancy, placental vascular growth, which is essential for supporting the rapidly growing fetus, is associated with marked elevations in blood flow. These vascular changes take place under chronic physiological low O2 (less than 2-8% O2 in human; chronic physiological normoxia, CPN) throughout pregnancy. O2 level below CPN pertinent to the placenta results in placental hypoxia. Such hypoxia can cause severe endothelial dysfunction, which is associated with adverse pregnancy outcomes (e.g., preeclampsia) and high risk of adult-onset cardiovascular diseases in children born to these pregnancy complications. However, our current knowledge about the mechanisms underlying fetoplacental endothelial function is derived primarily from cell models established under atmospheric O2 (~21% O2 at sea level, hyperoxia). Recent evidence has shown that fetoplacental endothelial cells cultured under CPN have distinct gene expression profiles and cellular responses compared with cells cultured under chronic hyperoxia. These data indicate the critical roles of CPN in programming fetal endothelial function and prompt us to re-examine the mechanisms governing fetoplacental endothelial function under CPN. Better understanding these mechanisms will facilitate us to develop preventive and therapeutic strategies for endothelial dysfunction-associated diseases (e.g., preeclampsia). This review will provide a brief summary on the impacts of CPN on endothelial function and its underlying mechanisms with a focus on fetoplacental endothelial cells.
Collapse
Affiliation(s)
- Chi Zhou
- Perinatal Research Laboratories, Department of Obstetrics and Gynecology, University of Wisconsin-Madison, Madison, Wisconsin
| | - Qing-Yun Zou
- Department of Vascular Surgery, First Affiliated Hospital, Harbin Medical University, Harbin, Heilongjiang, China
| | - Yi-Zhou Jiang
- Institute for Advanced Study, Shenzhen University, Shenzhen, Guangdong, China
| | - Jing Zheng
- Perinatal Research Laboratories, Department of Obstetrics and Gynecology, University of Wisconsin-Madison, Madison, Wisconsin.,Cardiovascular Medicine Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, China
| |
Collapse
|
25
|
Wijaya A, Maruf A, Wu W, Wang G. Recent advances in micro- and nano-bubbles for atherosclerosis applications. Biomater Sci 2020; 8:4920-4939. [DOI: 10.1039/d0bm00762e] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Micro- and nano-bubbles have been developed as powerful multimodal theranostic agents for atherosclerosis treatment.
Collapse
Affiliation(s)
- Andy Wijaya
- Key Laboratory for Biorheological Science and Technology of Ministry of Education
- State and Local Joint Engineering Laboratory for Vascular Implants
- Bioengineering College
- Faculty of Medicine
- Chongqing University
| | - Ali Maruf
- Key Laboratory for Biorheological Science and Technology of Ministry of Education
- State and Local Joint Engineering Laboratory for Vascular Implants
- Bioengineering College
- Faculty of Medicine
- Chongqing University
| | - Wei Wu
- Key Laboratory for Biorheological Science and Technology of Ministry of Education
- State and Local Joint Engineering Laboratory for Vascular Implants
- Bioengineering College
- Faculty of Medicine
- Chongqing University
| | - Guixue Wang
- Key Laboratory for Biorheological Science and Technology of Ministry of Education
- State and Local Joint Engineering Laboratory for Vascular Implants
- Bioengineering College
- Faculty of Medicine
- Chongqing University
| |
Collapse
|
26
|
Effect of magnesium-degradation products and hypoxia on the angiogenesis of human umbilical vein endothelial cells. Acta Biomater 2019; 98:269-283. [PMID: 30794987 DOI: 10.1016/j.actbio.2019.02.018] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2018] [Revised: 02/11/2019] [Accepted: 02/12/2019] [Indexed: 01/06/2023]
Abstract
Biodegradable magnesium (Mg) metals have been applied in orthopaedic and stent applications due to their biodegradability, bioabsorbability and adaptability to tissue regeneration. However, further investigations are still needed to understand how angiogenesis will respond to high concentrations of Mg and oxygen content differences, which are vital to vascular remodelling and bone fracture regeneration or tissue healing. Human primary endothelial cells were exposed to various concentrations (2-8 mM) of extracellular Mg degradation products under either hypoxia or normoxia. Increased proliferation was measured with Mg extracts under hypoxia but not under normoxia. Under normoxia and with Mg extracts, HUVEC migration exhibited a bell-shaped curve. The same pattern was observed with VEGFB expression, while VEGFA was constantly downregulated. Under hypoxia, migration and VEGFA levels remained constant; however, VEGFB was upregulated. Similarly, under normoxia, tube formation as well as VEGFA and VEGFB levels were downregulated. Nevertheless, under hypoxia, tube formation remained constant while VEGFA and VEGFB levels were upregulated. These results suggest that Mg extracts did not interfere with angiogenesis under hypoxia. STATEMENT OF SIGNIFICANCE: Neoangiogenesis, mediated by (e.g.) hypoxia, is a key factor for proper tissue healing Thus, effect of Mg degradation products under either hypoxia or normoxia on angiogenesis were investigated. Under normoxia and increased Mg concentrations, a general negative effect was measured on early (migration) and late (tubulogenesis) angiogenesis. However, under hypoxia, this effect was abolished. As magnesium degradation is an oxygen-dependant process, hypoxia condition may be a relevant factor to test material cytocompatibility in vitro.
Collapse
|
27
|
Wang L, Ge H, Peng L, Wang B. A meta-analysis of the relationship between VEGFR2 polymorphisms and atherosclerotic cardiovascular diseases. Clin Cardiol 2019; 42:860-865. [PMID: 31339592 PMCID: PMC6788482 DOI: 10.1002/clc.23233] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/02/2019] [Accepted: 07/08/2019] [Indexed: 01/11/2023] Open
Abstract
BACKGROUND Some previous studies explored associations between vascular endothelial growth factor receptor 2 (VEGFR2) polymorphisms and atherosclerotic cardiovascular diseases (ASCVD), with conflicting findings. HYPOTHESIS We thought that VEGFR2 polymorphisms may influence susceptibility to ASCVD. Here, we aimed to better analyze the relationship between VEGFR2 polymorphisms and ASCVD in a larger combined population by performing a meta-analysis. METHODS We searched Pubmed, Embase, and Web of Science for related articles. We calculated odds ratio (OR) and 95% confidence interval (CI) to estimate whether there are genetic associations between VEGFR2 polymorphisms and ASCVD. RESULTS Ten studies were included for this meta-analysis (5474 cases and 8584 controls). VEGFR2 rs1870377 (dominant comparison: 0.81 (0.73-0.89), I2 = 56%; recessive comparison: 1.40 (1.25-1.57), I2 = 34%; allele comparison: 0.81 (0.76-0.87), I2 = 0%), rs2071559 (dominant comparison: 0.83 (0.76-0.91), I2 = 0%; recessive comparison: 1.22 (1.07-1.38), I2 = 0%; allele comparison: 0.86 (0.81-0.92), I2 = 0%) and rs2305948 (dominant comparison: 0.79 (0.72-0.87), I2 = 25%; recessive comparison: 1.44 (1.08-1.92), I2 = 60%; allele comparison: 0.79 (0.68-0.92), I2 = 73%) polymorphisms were all found to be significantly associated with susceptibility to ASCVD in general population. Subgroup analyses by type of disease revealed similar significant findings for rs1870377, rs2071559, and rs2305948 polymorphisms in coronary artery disease (CAD) subgroup. Besides, positive results were also found for rs1870377 polymorphism in ischemic stroke (IS) subgroup. CONCLUSIONS In summary, this meta-analysis proved that these VEGFR2 polymorphisms could be used to identify individual with elevated susceptibility to ASCVD.
Collapse
Affiliation(s)
- Li Wang
- Department of HealthcareFirst Affiliated Hospital, Sun Yat‐sen UniversityGuangzhouChina
| | - Hui Ge
- Department of HealthcareFirst Affiliated Hospital, Sun Yat‐sen UniversityGuangzhouChina
| | - Longyun Peng
- Department of CardiologyFirst Affiliated Hospital, Sun Yat‐sen UniversityGuangzhouChina
| | - Bing Wang
- Department of RadiologySecond Affiliated Hospital, Army Medical UniversityChongqingChina
| |
Collapse
|
28
|
Dong G, Lin XH, Liu HH, Gao DM, Cui JF, Ren ZG, Chen RX. Intermittent hypoxia alleviates increased VEGF and pro-angiogenic potential in liver cancer cells. Oncol Lett 2019; 18:1831-1839. [PMID: 31423251 PMCID: PMC6607353 DOI: 10.3892/ol.2019.10486] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2018] [Accepted: 04/25/2019] [Indexed: 12/22/2022] Open
Abstract
Vascular endothelial growth factor (VEGF) is an important angiogenic factor. The VEGF rebound induced by hypoxia following transarterial embolization/chemoembolization for primary liver cancer is associated with treatment failure and poor survival rates in patients. The present study investigated the ability of intermittent hypoxia to alleviate the acute hypoxia-induced increase of VEGF and decrease the pro-angiogenic potential of liver cancer cells. The liver cancer cells were exposed to normoxia, or acute or intermittent hypoxia, and the expression of VEGF was determined using reverse transcription-quantitative polymerase chain reaction analysis and western blotting. The pro-angiogenic effects of acute or intermittent hypoxia-exposed liver cancer cells on endothelial cells were assessed in vitro and in vivo. The expression of VEGF in the liver cancer cells exposed to intermittent hypoxia was significantly lower than that in cells exposed to acute hypoxia. Compared with conditioned medium (CM) from acute hypoxia-exposed liver cancer cells, the CM from intermittent hypoxia-exposed liver cancer cells showed markedly less promotion of proliferation and tube formation in endothelial cells. Activation of the reactive oxygen species (ROS)/NF-κB/hypoxia-inducible factor-1α/VEGF signaling pathway was increased in the liver cancer cells exposed to acute hypoxia. Exposure to ROS scavenger N-acetyl-cysteine or NF-κB inhibitor PDTC inhibited the activation of the above pathway and the expression of VEGF induced by acute hypoxia. The in vivo pro-angiogenic effects of intermittent hypoxia-exposed liver cancer cells on endothelial cells were significantly reduced compared with those of acute hypoxia-exposed liver cancer cells. Intermittent hypoxia may alleviate the acute hypoxia-induced increase of VEGF and decrease the pro-angiogenic potential of liver cancer cells, suggesting a novel treatment strategy.
Collapse
Affiliation(s)
- Gang Dong
- Liver Cancer Institute, Zhongshan Hospital, Fudan University and Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Shanghai 200032, P.R. China
| | - Xia-Hui Lin
- Liver Cancer Institute, Zhongshan Hospital, Fudan University and Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Shanghai 200032, P.R. China
| | - Hua-Hua Liu
- Liver Cancer Institute, Zhongshan Hospital, Fudan University and Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Shanghai 200032, P.R. China
| | - Dong-Mei Gao
- Liver Cancer Institute, Zhongshan Hospital, Fudan University and Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Shanghai 200032, P.R. China
| | - Jie-Feng Cui
- Liver Cancer Institute, Zhongshan Hospital, Fudan University and Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Shanghai 200032, P.R. China
| | - Zheng-Gang Ren
- Liver Cancer Institute, Zhongshan Hospital, Fudan University and Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Shanghai 200032, P.R. China
| | - Rong-Xin Chen
- Liver Cancer Institute, Zhongshan Hospital, Fudan University and Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Shanghai 200032, P.R. China
| |
Collapse
|
29
|
Mansour SG, Zhang WR, Moledina DG, Coca SG, Jia Y, Thiessen-Philbrook H, McArthur E, Inoue K, Koyner JL, Shlipak MG, Wilson FP, Garg AX, Ishibe S, Parikh CR. The Association of Angiogenesis Markers With Acute Kidney Injury and Mortality After Cardiac Surgery. Am J Kidney Dis 2019; 74:36-46. [PMID: 30955944 DOI: 10.1053/j.ajkd.2019.01.028] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2018] [Accepted: 01/26/2019] [Indexed: 11/11/2022]
Abstract
RATIONALE & OBJECTIVE The process of angiogenesis after kidney injury may determine recovery and long-term outcomes. We evaluated the association of angiogenesis markers with acute kidney injury (AKI) and mortality after cardiac surgery. STUDY DESIGN Prospective cohort. SETTING & PARTICIPANTS 1,444 adults undergoing cardiac surgery in the TRIBE-AKI (Translational Research Investigating Biomarker Endpoints for Acute Kidney Injury) cohort. EXPOSURES Plasma concentrations of 2 proangiogenic markers (vascular endothelial growth factor A [VEGF] and placental growth factor [PGF]) and 1 antiangiogenic marker (soluble VEGF receptor 1 [VEGFR1]), measured pre- and postoperatively within 6 hours after surgery. OUTCOMES AKI, long AKI duration (≥7 days), and 1-year all-cause mortality. ANALYTICAL APPROACH Multivariable logistic regression. RESULTS Following cardiac surgery, plasma VEGF concentrations decreased 2-fold, and PGF and VEGFR1 concentrations increased 1.5- and 8-fold, respectively. There were no meaningful associations of preoperative concentrations of angiogenic markers with outcomes of AKI and mortality. Higher postoperative VEGF and PGF concentrations were independently associated with lower odds of AKI (adjusted ORs of 0.89 [95% CI, 0.82-0.98] and 0.69 [95% CI, 0.55-0.87], respectively), long AKI duration (0.65 [95% CI, 0.49-0.87] and 0.48 [95% CI, 0.28-0.82], respectively), and mortality (0.74 [95% CI, 0.62-0.89] and 0.46 [95% CI, 0.31-0.68], respectively). In contrast, higher postoperative VEGFR1 concentrations were independently associated with higher odds of AKI (1.56; 95% CI, 1.31-1.87), long AKI duration (1.75; 95% CI, 1.09-2.82), and mortality (2.28; 95% CI, 1.61-3.22). LIMITATIONS Angiogenesis markers were not measured after hospital discharge, so we were unable to determine long-term trajectories of angiogenesis marker levels during recovery and follow-up. CONCLUSIONS Higher levels of postoperative proangiogenic markers, VEGF and PGF, were associated with lower AKI and mortality risk, whereas higher postoperative antiangiogenic VEGFR1 levels were associated with higher risk for AKI and mortality.
Collapse
Affiliation(s)
- Sherry G Mansour
- Program of Applied Translational Research, Department of Medicine, Yale University School of Medicine, New Haven, CT; Section of Nephrology, Yale University School of Medicine, New Haven, CT
| | - William R Zhang
- Program of Applied Translational Research, Department of Medicine, Yale University School of Medicine, New Haven, CT; Kidney Health Research Collaborative and Division of General Internal Medicine, San Francisco Veterans Affairs Medical Center, University of California, San Francisco, CA
| | - Dennis G Moledina
- Program of Applied Translational Research, Department of Medicine, Yale University School of Medicine, New Haven, CT; Section of Nephrology, Yale University School of Medicine, New Haven, CT
| | - Steven G Coca
- Division of Nephrology, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Yaqi Jia
- Division of Nephrology, Department of Medicine, Johns Hopkins University, Baltimore, MD
| | | | - Eric McArthur
- Institute for Clinical Evaluative Sciences, London, Ontario, Canada
| | - Kazunori Inoue
- Section of Nephrology, Yale University School of Medicine, New Haven, CT
| | - Jay L Koyner
- Section of Nephrology, University of Chicago, Chicago, IL
| | - Michael G Shlipak
- Kidney Health Research Collaborative and Division of General Internal Medicine, San Francisco Veterans Affairs Medical Center, University of California, San Francisco, CA
| | - F Perry Wilson
- Program of Applied Translational Research, Department of Medicine, Yale University School of Medicine, New Haven, CT; Section of Nephrology, Yale University School of Medicine, New Haven, CT
| | - Amit X Garg
- Department of Health Research Methods, Evidence, and Impact, McMaster University, Hamilton, Ontario, Canada
| | - Shuta Ishibe
- Section of Nephrology, Yale University School of Medicine, New Haven, CT
| | - Chirag R Parikh
- Division of Nephrology, Department of Medicine, Johns Hopkins University, Baltimore, MD.
| | | |
Collapse
|
30
|
Kozinszky Z, Altorjay A, Molnar A, Nyári T, Vari SG, Nemeth G, Suranyi A. Ultrasonographic Evaluation of Glycemic Control Effect on Placental Vascularization in Pregnancy with Type 1 Diabetes Mellitus. Exp Clin Endocrinol Diabetes 2019; 128:788-795. [PMID: 30641608 DOI: 10.1055/a-0824-6362] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
AIMS The aim of our study was to assess the effect of glycemic control on placental vascularization in pregnancies complicated by type 1 diabetes mellitus (T1DM) and to compare dataset of optimal/suboptimal glycemic control to normal placental 3-dimensional power Doppler (3-DPD) indices in 2nd and 3rd trimester. METHODS Placental vascularization of pregnant women was prospectively evaluated by 3-DPD ((vascularization-index (VI); flow-index (FI); vascularization-flow-index (VFI)) ultrasound technique. The normal pregnancies (n=214) were compared to those complicated by T1DM (n=53) with optimal (HbA1C≤6%;≤ 42 mmol/mol) and suboptimal (HbA1C>6%;>42 mmol/mol) glycemic control. RESULTS Pregnancies complicated by T1DM expressed lower placental vascularization indices as compared with normal pregnancies (adjusted odds ratio (AOR) for VI:0.86; FI:0.94; VFI:0.76). Placental 3-DPD indices have a significant correlation with HbA1C and optimal glycemic control is associated with lower placental perfusion (AOR for VI:1.64; FI:1.13; VFI:2.34). Short-term adverse neonatal outcome was predicted by lower 3-DPD indices (AORVI:0.83, AORFI:0.93, AORVFI:0.66, p<0.05 for each index). Besides the glycemic control, the pregestational body mass index (BMI), had significant influences on placental perfusion. CONCLUSIONS VI displayed the best screening ability for suboptimal glycemic control with a sensitivity of 90.9%. The suboptimal glycemic control has a direct deteriorating effect on placental vasculature. Therefore the ultrasound examination could be an adjunct diagnostic modality for pregnant women with T1DM.
Collapse
Affiliation(s)
- Zoltan Kozinszky
- Department of Obstetrics and Gynecology, Blekinge Hospital, Karlskrona, Sweden
| | - AbelT Altorjay
- Department of Obstetrics and Gynecology, University of Szeged, Szeged, Hungary
| | - Andras Molnar
- Department of Obstetrics and Gynecology, University of Szeged, Szeged, Hungary
| | - Tibor Nyári
- Department of Medical Physics and Informatics, University of Szeged, Szeged, Hungary
| | - Sandor G Vari
- International Research and Innovation in Medicine Program, Cedars - Sinai Medical Center, Los Angeles, CA, USA
| | - Gabor Nemeth
- Department of Obstetrics and Gynecology, University of Szeged, Szeged, Hungary
| | - Andrea Suranyi
- Department of Obstetrics and Gynecology, University of Szeged, Szeged, Hungary
| |
Collapse
|
31
|
Billaud M, Hill JC, Richards TD, Gleason TG, Phillippi JA. Medial Hypoxia and Adventitial Vasa Vasorum Remodeling in Human Ascending Aortic Aneurysm. Front Cardiovasc Med 2018; 5:124. [PMID: 30276199 PMCID: PMC6151311 DOI: 10.3389/fcvm.2018.00124] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2018] [Accepted: 08/20/2018] [Indexed: 11/29/2022] Open
Abstract
Human ascending aortic aneurysms characteristically exhibit cystic medial degeneration of the aortic wall encompassing elastin degeneration, proteoglycan accumulation and smooth muscle cell loss. Most studies have focused on the aortic media and there is a limited understanding of the importance of the adventitial layer in the setting of human aneurysmal disease. We recently demonstrated that the adventitial ECM contains key angiogenic factors that are downregulated in aneurysmal aortic specimens. In this study, we investigated the adventitial microvascular network (vasa vasorum) of aneurysmal aortic specimens of different etiology and hypothesized that the vasa vasorum is disrupted in patients with ascending aortic aneurysm. Morphometric analyses of hematoxylin and eosin-stained human aortic cross-sections revealed evidence of vasa vasorum remodeling in aneurysmal specimens, including reduced density of vessels, increased lumen area and thickening of smooth muscle actin-positive layers. These alterations were inconsistently observed in specimens of bicuspid aortic valve (BAV)-associated aortopathy, while vasa vasorum remodeling was typically observed in aneurysms arising in patients with the morphologically normal tricuspid aortic valve (TAV). Gene expression of hypoxia-inducible factor 1α and its downstream targets, metallothionein 1A and the pro-angiogenic factor vascular endothelial growth factor, were down-regulated in the adventitia of aneurysmal specimens when compared with non-aneurysmal specimens, while the level of the anti-angiogenic factor thrombospondin-1 was elevated. Immunodetection of glucose transporter 1 (GLUT1), a marker of chronic tissue hypoxia, was minimal in non-aneurysmal medial specimens, and locally accumulated within regions of elastin degeneration, particularly in TAV-associated aneurysms. Quantification of GLUT1 revealed elevated levels in the aortic media of TAV-associated aneurysms when compared to non-aneurysmal counterparts. We detected evidence of chronic inflammation as infiltration of lymphoplasmacytic cells in aneurysmal specimens, with a higher prevalence of lymphoplasmacytic infiltrates in aneurysmal specimens from patients with TAV compared to that of patients with BAV. These data highlight differences in vasa vasorum remodeling and associated medial chronic hypoxia markers between aneurysms of different etiology. These aberrations could contribute to malnourishment of the aortic media and could conceivably participate in the pathogenesis of thoracic aortic aneurysm.
Collapse
Affiliation(s)
- Marie Billaud
- Department of Cardiothoracic Surgery, University of Pittsburgh, Pittsburgh, PA, United States.,McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA, United States.,Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, United States
| | - Jennifer C Hill
- Department of Cardiothoracic Surgery, University of Pittsburgh, Pittsburgh, PA, United States
| | - Tara D Richards
- Department of Cardiothoracic Surgery, University of Pittsburgh, Pittsburgh, PA, United States
| | - Thomas G Gleason
- Department of Cardiothoracic Surgery, University of Pittsburgh, Pittsburgh, PA, United States.,McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA, United States.,Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, United States.,Center for Vascular Remodeling and Regeneration, University of Pittsburgh, Pittsburgh, PA, United States
| | - Julie A Phillippi
- Department of Cardiothoracic Surgery, University of Pittsburgh, Pittsburgh, PA, United States.,McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA, United States.,Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, United States.,Center for Vascular Remodeling and Regeneration, University of Pittsburgh, Pittsburgh, PA, United States
| |
Collapse
|
32
|
Zhao X, Meng L, Jiang J, Wu X. Vascular endothelial growth factor gene polymorphisms and coronary heart disease: a systematic review and meta-analysis. Growth Factors 2018; 36:153-163. [PMID: 30317903 DOI: 10.1080/08977194.2018.1477141] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
We performed this study to better elucidate the relationship between vascular endothelial growth factor (VEGF) polymorphisms and coronary heart disease (CHD). Eligible articles were searched in PubMed, Medline, Embase, Scopus and CNKI. A total of 24 studies containing 6489 CHD patients and 5664 control subjects were analyzed. Our overall and subgroup analyses suggested that rs699947 polymorphism was significantly associated with CHD susceptibility in both Caucasians and Asians, rs1570360 polymorphism was significantly associated with CHD susceptibility in Caucasians, and rs3025039 polymorphism was significantly associated with CHD susceptibility in Asians. Besides, rs3025039 polymorphism was significantly correlated with the number of affected coronary arteries, while rs699947 and rs2010963 polymorphisms were significantly correlated with poor collateral circulation in CHD patients. Overall, our findings indicate that VEGF rs699947, rs1570360, and rs3025039 polymorphisms may affect CHD susceptibility. Moreover, VEGF rs699947 and rs2010963 polymorphisms may serve as genetic biomarkers of poor collateral circulation after myocardial ischemia.
Collapse
Affiliation(s)
- Xiangwen Zhao
- a Cardiovascular Inpatient Ward 2, Lanling People's Hospital , Linyi , Shandong , China
| | - Liang Meng
- b Research Center Laboratory , Lanling People's Hospital , Linyi , Shandong , China
| | - Jimin Jiang
- b Research Center Laboratory , Lanling People's Hospital , Linyi , Shandong , China
| | - Xinglong Wu
- b Research Center Laboratory , Lanling People's Hospital , Linyi , Shandong , China
| |
Collapse
|
33
|
Lai XM, Liu SY, Tsai YT, Sun GH, Chang SY, Huang SM, Cha TL. HAF mediates the evasive resistance of anti-angiogenesis TKI through disrupting HIF-1α and HIF-2α balance in renal cell carcinoma. Oncotarget 2018; 8:49713-49724. [PMID: 28572533 PMCID: PMC5564801 DOI: 10.18632/oncotarget.17923] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2017] [Accepted: 05/03/2017] [Indexed: 12/29/2022] Open
Abstract
Anti-angiogenesis has emerged as a standard of care for metastatic renal cell carcinoma. However, long-lasting efficacy is seldom reached, and evasive resistance eventually occurs under anti-angiogenic tyrosine kinase inhibitor (TKI) therapy. To establish new therapeutic strategies, investigating the molecular mechanism of resistance is critically important. In our study, human umbilical vascular endothelial cells (HUVECs) were incubated with TKI treatment in conditioned medium derived from renal cancer cells (RCCs) to demonstrate cell viability. Quantitative real time PCR or Western blotting analysis detected the fluctuation of transcriptional factors HIF-1α and HIF-2α in RCCs under TKI treatment. We demonstrated the alteration of a specific cytokine produced from RCCs under normoxia or hypoxia incubation by utilizing a cytokine RT-PCR primer array. We found that the anti-angiogenic TKI sunitinib disrupted the balance between HIF-1α and HIF-2α in RCCs and led to a protective effect on HUVECs against sunitinib treatment when cultured with conditioned medium. Mechanistically, RCCs treated with sunitinib resulted in down-regulation of HIF-1α, but not HIF-2α, through reduction of both mRNA and protein levels. The down-regulation of HIF-1α by sunitinib occurred via hypoxia associated factor (HAF), which also enhanced HIF-2α transactivation activity to increase the production of pro-angiogenic factors and cytokines and promote HUVEC proliferation. This phenomenon was observed in ACHN and A498 cells, which express both HIF-1α and HIF-2α, but was not observed in 786-O cells, which express only HIF-2α. Our results illustrated that targeting both angiogenesis and hypoxia pathways might provide a resolution to dealing with the devastating effects of anti-angiogenesis resistance.
Collapse
Affiliation(s)
- Xiang-Me Lai
- Graduate Institute of Life Sciences, National Defense Medical Center, Taipei, Taiwan, R.O.C
| | - Shu-Yu Liu
- Division of Urology, Department of Surgery, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan, R.O.C
| | - Yi-Ta Tsai
- Graduate Institute of Medical Sciences, National Defense Medical Center, Taipei, Taiwan, R.O.C
| | - Guang-Huan Sun
- Division of Urology, Department of Surgery, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan, R.O.C
| | - Sun-Yran Chang
- Division of Urology, Department of Surgery, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan, R.O.C.,Buddhist Tzu Chi General Hospital, Taipei, Taiwan, R.O.C
| | - Shih-Ming Huang
- Graduate Institute of Life Sciences, National Defense Medical Center, Taipei, Taiwan, R.O.C.,Graduate Institute of Medical Sciences, National Defense Medical Center, Taipei, Taiwan, R.O.C.,Department of Biochemistry, National Defense Medical Center, Taipei, Taiwan, R.O.C
| | - Tai-Lung Cha
- Graduate Institute of Life Sciences, National Defense Medical Center, Taipei, Taiwan, R.O.C.,Division of Urology, Department of Surgery, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan, R.O.C.,Graduate Institute of Medical Sciences, National Defense Medical Center, Taipei, Taiwan, R.O.C.,Department of Biochemistry, National Defense Medical Center, Taipei, Taiwan, R.O.C
| |
Collapse
|
34
|
Chronic kidney disease induces a systemic microangiopathy, tissue hypoxia and dysfunctional angiogenesis. Sci Rep 2018; 8:5317. [PMID: 29593228 PMCID: PMC5871820 DOI: 10.1038/s41598-018-23663-1] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2017] [Accepted: 03/19/2018] [Indexed: 12/21/2022] Open
Abstract
Chronic kidney disease (CKD) is associated with excessive mortality from cardiovascular disease (CVD). Endothelial dysfunction, an early manifestation of CVD, is consistently observed in CKD patients and might be linked to structural defects of the microcirculation including microvascular rarefaction. However, patterns of microvascular rarefaction in CKD and their relation to functional deficits in perfusion and oxygen delivery are currently unknown. In this in-vivo microscopy study of the cremaster muscle microcirculation in BALB/c mice with moderate to severe uremia, we show in two experimental models (adenine feeding or subtotal nephrectomy), that serum urea levels associate incrementally with a distinct microangiopathy. Structural changes were characterized by a heterogeneous pattern of focal microvascular rarefaction with loss of coherent microvascular networks resulting in large avascular areas. Corresponding microvascular dysfunction was evident by significantly diminished blood flow velocity, vascular tone, and oxygen uptake. Microvascular rarefaction in the cremaster muscle paralleled rarefaction in the myocardium, which was accompanied by a decrease in transcription levels not only of the transcriptional regulator HIF-1α, but also of its target genes Angpt-2, TIE-1 and TIE-2, Flkt-1 and MMP-9, indicating an impaired hypoxia-driven angiogenesis. Thus, experimental uremia in mice associates with systemic microvascular disease with rarefaction, tissue hypoxia and dysfunctional angiogenesis.
Collapse
|
35
|
Abstract
Renovascular disease (RVD), which is prevalent in the elderly, significantly increases cardiovascular risk and can progressively deteriorate renal function. The loss of renal function in patients with RVD is associated with a progressive dysfunction, damage, and loss of renal microvessels, which can be combined with decreased renal bioavailability of vascular endothelial growth factor (VEGF) and a defective vascular repair and proliferation. This association has been the impetus for recent efforts that have focused on developing methods to stop the progression of renal injury by protecting the renal microvasculature. This mini-review focuses on recent studies supporting potential applications of VEGF therapy for the kidney and discusses underlying mechanisms of renoprotection.
Collapse
Affiliation(s)
- Erika Guise
- Department of Physiology and Biophysics, University of Mississippi Medical Center , Jackson, Mississippi
| | - Alejandro R Chade
- Department of Physiology and Biophysics, University of Mississippi Medical Center , Jackson, Mississippi.,Department of Medicine, University of Mississippi Medical Center , Jackson, Mississippi.,Department of Radiology, University of Mississippi Medical Center , Jackson, Mississippi
| |
Collapse
|
36
|
Cannizzo CM, Adonopulos AA, Solly EL, Ridiandries A, Vanags LZ, Mulangala J, Yuen SCG, Tsatralis T, Henriquez R, Robertson S, Nicholls SJ, Di Bartolo BA, Ng MKC, Lam YT, Bursill CA, Tan JTM. VEGFR2 is activated by high-density lipoproteins and plays a key role in the proangiogenic action of HDL in ischemia. FASEB J 2018; 32:2911-2922. [PMID: 29401597 DOI: 10.1096/fj.201700617r] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
High-density lipoproteins augment hypoxia-induced angiogenesis by inducing the key angiogenic vascular endothelial growth factor A (VEGFA) and total protein levels of its receptor 2 (VEGFR2). The activation/phosphorylation of VEGFR2 is critical for mediating downstream, angiogenic signaling events. This study aimed to determine whether reconstituted high-density lipoprotein (rHDL) activates VEGFR2 phosphorylation and the downstream signaling events and the importance of VEGFR2 in the proangiogenic effects of rHDL in hypoxia. In vitro, rHDL increased VEGFR2 activation and enhanced phosphorylation of downstream, angiogenic signaling proteins ERK1/2 and p38 MAPK in hypoxia. Incubation with a VEGFR2-neutralizing antibody attenuated rHDL-induced phosphorylation of VEGFR2, ERK1/2, p38 MAPK, and tubule formation. In a murine model of ischemia-driven neovascularization, rHDL infusions enhanced blood perfusion and augmented capillary and arteriolar density. Infusion of a VEGFR2-neutralizing antibody ablated those proangiogenic effects of rHDL. Circulating Sca1+/CXCR4+ angiogenic progenitor cell levels, important for neovascularization in response to ischemia, were higher in rHDL-infused mice 3 d after ischemic induction, but that did not occur in mice that also received the VEGFR2-neutralizing antibody. In summary, VEGFR2 has a key role in the proangiogenic effects of rHDL in hypoxia/ischemia. These findings have therapeutic implications for angiogenic diseases associated with an impaired response to tissue ischemia.-Cannizzo, C. M., Adonopulos, A. A., Solly, E. L., Ridiandries, A., Vanags, L. Z., Mulangala, J., Yuen, S. C. G., Tsatralis, T., Henriquez, R., Robertson, S., Nicholls, S. J., Di Bartolo, B. A., Ng, M. K. C., Lam, Y. T., Bursill, C. A., Tan, J. T. M. VEGFR2 is activated by high-density lipoproteins and plays a key role in the proangiogenic action of HDL in ischemia.
Collapse
Affiliation(s)
- Carla M Cannizzo
- The Heart Research Institute, Newtown, New South Wales, Australia.,Sydney Medical School, The University of Sydney, Camperdown, New South Wales, Australia
| | - Aaron A Adonopulos
- The Heart Research Institute, Newtown, New South Wales, Australia.,Sydney Medical School, The University of Sydney, Camperdown, New South Wales, Australia
| | - Emma L Solly
- Heart Health Theme, South Australian Health and Medical Research Institute, Adelaide, South Australia, Australia
| | - Anisyah Ridiandries
- The Heart Research Institute, Newtown, New South Wales, Australia.,Sydney Medical School, The University of Sydney, Camperdown, New South Wales, Australia
| | - Laura Z Vanags
- The Heart Research Institute, Newtown, New South Wales, Australia.,Sydney Medical School, The University of Sydney, Camperdown, New South Wales, Australia
| | - Jocelyne Mulangala
- Heart Health Theme, South Australian Health and Medical Research Institute, Adelaide, South Australia, Australia.,Adelaide Medical School, Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide, South Australia, Australia; and
| | - Sui Ching G Yuen
- The Heart Research Institute, Newtown, New South Wales, Australia.,Sydney Medical School, The University of Sydney, Camperdown, New South Wales, Australia
| | - Tania Tsatralis
- The Heart Research Institute, Newtown, New South Wales, Australia
| | - Rodney Henriquez
- The Heart Research Institute, Newtown, New South Wales, Australia
| | - Stacy Robertson
- The Heart Research Institute, Newtown, New South Wales, Australia.,Sydney Medical School, The University of Sydney, Camperdown, New South Wales, Australia
| | - Stephen J Nicholls
- Heart Health Theme, South Australian Health and Medical Research Institute, Adelaide, South Australia, Australia.,Adelaide Medical School, Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide, South Australia, Australia; and
| | - Belinda A Di Bartolo
- Heart Health Theme, South Australian Health and Medical Research Institute, Adelaide, South Australia, Australia.,Adelaide Medical School, Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide, South Australia, Australia; and
| | - Martin K C Ng
- The Heart Research Institute, Newtown, New South Wales, Australia.,Sydney Medical School, The University of Sydney, Camperdown, New South Wales, Australia.,Department of Cardiology, Royal Prince Alfred Hospital, Camperdown, New South Wales, Australia
| | - Yuen Ting Lam
- The Heart Research Institute, Newtown, New South Wales, Australia.,Sydney Medical School, The University of Sydney, Camperdown, New South Wales, Australia
| | - Christina A Bursill
- The Heart Research Institute, Newtown, New South Wales, Australia.,Sydney Medical School, The University of Sydney, Camperdown, New South Wales, Australia.,Heart Health Theme, South Australian Health and Medical Research Institute, Adelaide, South Australia, Australia.,Adelaide Medical School, Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide, South Australia, Australia; and
| | - Joanne T M Tan
- The Heart Research Institute, Newtown, New South Wales, Australia.,Sydney Medical School, The University of Sydney, Camperdown, New South Wales, Australia.,Heart Health Theme, South Australian Health and Medical Research Institute, Adelaide, South Australia, Australia.,Adelaide Medical School, Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide, South Australia, Australia; and
| |
Collapse
|
37
|
Long L, Qiu H, Cai B, Chen N, Lu X, Zheng S, Ye X, Li Y. Hyperglycemia induced testicular damage in type 2 diabetes mellitus rats exhibiting microcirculation impairments associated with vascular endothelial growth factor decreased via PI3K/Akt pathway. Oncotarget 2018; 9:5321-5336. [PMID: 29435181 PMCID: PMC5797052 DOI: 10.18632/oncotarget.23915] [Citation(s) in RCA: 64] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2017] [Accepted: 12/01/2017] [Indexed: 12/13/2022] Open
Abstract
As an endocrine disease, type 2 diabetes mellitus (T2DM) can cause testicular damage which induces male infertility. However, the underlying mechanism is still not clear. We prove that T2DM induced testicular microcirculation impairment involves the decrease of VEGF and these actions are regulated by PI3K/Akt pathway. In our study, rats were divided into three groups (n=8): control group, diabetes group and diabetes + VEGF group. Intraperitoneal injection of streptozotocin (STZ, 65mg/Kg, at 9th week) and daily high-fat diet were used to establish T2DM rat model. Serum glucose in diabetes group and diabetes + VEGF group obviously exceeded 13mmol/L after STZ injection. Immunohistochemical studies indicated that VEGF level in diabetes group significantly decreased. In diabetes group, testicular blood velocity and vascular area reduced evaluated by Doppler and FITC. Furthermore, atrophic testicular morphology and increasing apoptosis cells were evaluated by haematoxylin and eosin staining and TUNEL assay. In diabetes + VEGF group, the administration of VEGF (intraperitoneally, 10mg/kg) can significantly alleviated hyperglycemia-induced impairment of testes in above aspects. Finally, we used Western blot to analyze the mechanism of hyperglycemia-induced testicular VEGF decrease. The results indicated that hyperglycemia-induced VEGF decreased is regulated by PI3K/Akt pathway in Rats testicular sertoli cells (RTSCs). Together, we demonstrate that T2DM can reduce testicular VEGF expression, which results in testicular microcirculation impairment, and then induces testicular morphological disarrangement and functional disorder. These actions are triggered by PI3K/Akt pathway. Our findings provide solid evidence for VEGF becoming a therapeutic target in T2DM related male infertility.
Collapse
Affiliation(s)
- Lingli Long
- Translation Medicine Center, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Han Qiu
- Department of Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Bing Cai
- The Reproductive Center of the First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Ningning Chen
- Department of Orthopedics, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| | - Xiaofang Lu
- Department of Pathology, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Shuhui Zheng
- Translation Medicine Center, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Xiaoxin Ye
- University of New South Wales, Sydney, Australia
| | - Yubin Li
- The Reproductive Center of the First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| |
Collapse
|
38
|
Abstract
Vascular endothelial growth factor (VEGF) is a potent angiogenic factor. Despite upregulation of VEGF in the brain in Alzheimer's disease (AD), probably in response to amyloid-β, vasoconstriction, and tissue hypoxia, there is no consequent increase in microvessel density. VEGF binds to and activates VEGF receptor 2 (VEGFR2), but also binds to VEGF receptor 1 (VEGFR1), which exists in less-active membrane-bound and inactive soluble (sVEGFR1) forms and inhibits pro-angiogenic signaling. We have investigated whether altered expression of VEGF receptors might account for the lack of angiogenic response to VEGF in AD. We assessed the cellular distribution and protein level of VEGFR1 and VEGFR2 in parietal cortex from 50 AD and 36 age-matched control brains, and related the findings to measurements of VEGF and von Willebrand factor level (a marker of microvessel density) in the same tissue samples. VEGFR2 was expressed by neurons, astrocytes and endothelial cells. VEGFR1 was expressed predominantly neuronally and was significantly reduced in AD (p = 0.02). Western blot analysis on a subset of brains showed reduction in VEGFR1:sVEGFR1 in AD (p = 0.046). The lack of angiogenesis despite cerebral hypoperfusion in AD is not explained by altered expression of VEGFR2 or total VEGFR1; indeed, the downregulation of VEGFR1 may represent a pro-angiogenic response to the hypoperfusion. However, the relative increase in sVEGFR1 would be expected to have an anti-angiogenic effect which may be a factor in AD.
Collapse
Affiliation(s)
- Rachel Harris
- Institute of Clinical Neurosciences, University of Bristol, School of Medicine, Level 2 Learning and Research, Southmead Hospital, Bristol, UK
| | - James Scott Miners
- Institute of Clinical Neurosciences, University of Bristol, School of Medicine, Level 2 Learning and Research, Southmead Hospital, Bristol, UK
| | - Shelley Allen
- Institute of Clinical Neurosciences, University of Bristol, School of Medicine, Level 2 Learning and Research, Southmead Hospital, Bristol, UK
| | - Seth Love
- Institute of Clinical Neurosciences, University of Bristol, School of Medicine, Level 2 Learning and Research, Southmead Hospital, Bristol, UK
| |
Collapse
|
39
|
Masum MA, Ichii O, Elewa YHA, Nakamura T, Kon Y. Local CD34-positive capillaries decrease in mouse models of kidney disease associating with the severity of glomerular and tubulointerstitial lesions. BMC Nephrol 2017; 18:280. [PMID: 28870174 PMCID: PMC5584339 DOI: 10.1186/s12882-017-0694-3] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2017] [Accepted: 08/21/2017] [Indexed: 01/06/2023] Open
Abstract
BACKGROUND The renal vasculature plays important roles in both homeostasis and pathology. In this study, we examined pathological changes in the renal microvascular in mouse models of kidney diseases. METHODS Glomerular lesions (GLs) in autoimmune disease-prone male BXSB/MpJ-Yaa (Yaa) mice and tubulointerstitial lesions (TILs) in male C57BL/6 mice subjected to unilateral ureteral obstruction (UUO) for 7 days were studied. Collected kidneys were examined using histopathological techniques. A nonparametric Mann-Whitney U test (P < 0.05) was performed to compare healthy controls and the experimental mice. The Kruskal-Wallis test was used to compare three or more groups, and multiple comparisons were performed using Scheffe's method when significant differences were observed (P < 0.05). RESULTS Yaa mice developed severe autoimmune glomerulonephritis, and the number of CD34+ glomerular capillaries decreased significantly in GLs compared to that in control mice. However, UUO-treated mice showed severe TILs only, and CD34+ tubulointerstitial capillaries were decreased significantly in TILs with the progression of tubulointerstitial fibrosis compared to those in untreated control kidneys. Infiltrations of B-cells, T-cells, and macrophages increased significantly in the respective lesions of both disease models (P < 0.05). In observations of vascular corrosion casts by scanning electron microscopy and of microfil rubber-perfused thick kidney sections by fluorescence microscopy, segmental absences of capillaries were observed in the GLs and TILs of Yaa and UUO-treated mice, respectively. Further, transmission electron microscopy revealed capillary endothelial injury in the respective lesions of both models. The numbers of CD34+ glomerular and tubulointerstitial capillaries were negatively correlated with all examined parameters in GLs (P < 0.05) and TILs (P < 0.01), respectively. CONCLUSIONS From the analysis of mouse models, we identified inverse pathological correlations between the number of local capillaries in GLs and TILs and the severity of kidney diseases.
Collapse
Affiliation(s)
- Md Abdul Masum
- Laboratory of Anatomy, Graduate School of Veterinary Medicine, Hokkaido University, Kita 18, Nishi 9, Kita-ku, Sapporo, Japan
| | - Osamu Ichii
- Laboratory of Anatomy, Graduate School of Veterinary Medicine, Hokkaido University, Kita 18, Nishi 9, Kita-ku, Sapporo, Japan
| | - Yaser Hosny Ali Elewa
- Laboratory of Anatomy, Graduate School of Veterinary Medicine, Hokkaido University, Kita 18, Nishi 9, Kita-ku, Sapporo, Japan
- Department of Histology, Faculty of Veterinary Medicine, Zagazig University, Zagazig, Egypt
| | - Teppei Nakamura
- Laboratory of Anatomy, Graduate School of Veterinary Medicine, Hokkaido University, Kita 18, Nishi 9, Kita-ku, Sapporo, Japan
- Section of Biological Safety Research, Chitose Laboratory, Japan Food Research Laboratories, Tokyo, Japan
| | - Yasuhiro Kon
- Laboratory of Anatomy, Graduate School of Veterinary Medicine, Hokkaido University, Kita 18, Nishi 9, Kita-ku, Sapporo, Japan
| |
Collapse
|
40
|
Keeley TP, Siow RCM, Jacob R, Mann GE. A PP2A-mediated feedback mechanism controls Ca 2+-dependent NO synthesis under physiological oxygen. FASEB J 2017; 31:5172-5183. [PMID: 28760745 PMCID: PMC5690389 DOI: 10.1096/fj.201700211r] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2017] [Accepted: 07/17/2017] [Indexed: 12/20/2022]
Abstract
Intracellular O2 is a key regulator of NO signaling, yet most in vitro studies are conducted in atmospheric O2 levels, hyperoxic with respect to the physiologic milieu. We investigated NO signaling in endothelial cells cultured in physiologic (5%) O2 and stimulated with histamine or shear stress. Culture of cells in 5% O2 (>5 d) decreased histamine- but not shear stress–stimulated endothelial (e)NOS activity. Unlike cells adapted to a hypoxic environment (1% O2), those cultured in 5% O2 still mobilized sufficient Ca2+ to activate AMPK. Enhanced expression and membrane targeting of PP2A-C was observed in 5% O2, resulting in greater interaction with eNOS in response to histamine. Moreover, increased dephosphorylation of eNOS in 5% O2 was Ca2+-sensitive and reversed by okadaic acid or PP2A-C siRNA. The present findings establish that Ca2+ mobilization stimulates both NO synthesis and PP2A-mediated eNOS dephosphorylation, thus constituting a novel negative feedback mechanism regulating eNOS activity not present in response to shear stress. This, coupled with enhanced NO bioavailability, underpins differences in NO signaling induced by inflammatory and physiologic stimuli that are apparent only in physiologic O2 levels. Furthermore, an explicit delineation between physiologic normoxia and genuine hypoxia is defined here, with implications for our understanding of pathophysiological hypoxia.—Keeley, T. P., Siow, R. C. M., Jacob, R., Mann, G. E. A PP2A-mediated feedback mechanism controls Ca2+-dependent NO synthesis under physiological oxygen.
Collapse
Affiliation(s)
- Thomas P Keeley
- Cardiovascular Division, King's British Heart Foundation Centre of Research Excellence, Faculty of Life Sciences and Medicine, King's College London, London, United Kingdom
| | - Richard C M Siow
- Cardiovascular Division, King's British Heart Foundation Centre of Research Excellence, Faculty of Life Sciences and Medicine, King's College London, London, United Kingdom
| | - Ron Jacob
- Cardiovascular Division, King's British Heart Foundation Centre of Research Excellence, Faculty of Life Sciences and Medicine, King's College London, London, United Kingdom
| | - Giovanni E Mann
- Cardiovascular Division, King's British Heart Foundation Centre of Research Excellence, Faculty of Life Sciences and Medicine, King's College London, London, United Kingdom
| |
Collapse
|
41
|
Angiogenic and angiostatic factors in renal scleroderma-associated vasculopathy. Microvasc Res 2017; 114:41-45. [PMID: 28602918 DOI: 10.1016/j.mvr.2017.06.003] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2017] [Revised: 06/07/2017] [Accepted: 06/07/2017] [Indexed: 01/08/2023]
Abstract
BACKGROUND The angiogenesis in systemic sclerosis (SSc) is impaired. An imbalance of pro-angiogenic factors and angiogenesis inhibitors has been implicated in the progression of peripheral microvascular damage, defective vascular repair and fibrosis. Intrarenal resistance index are considered markers of renal vasculopathy. The aim of the study is to evaluate angiogenic and angiostatic factors (VEGF and endostatin) in SSc patients and to correlate with intrarenal hemodynamic parameters. METHODS 91 SSc patients were enrolled in this study. Serum VEGF and endostatin levels were determined. All patients underwent a renal Doppler ultrasound RESULTS: A significant positive correlation was observed between endostatin and renal Doppler parameters (p<0.0001). A negative correlation was observed between serum levels of endostatin and eGFR (p<0.01). In SSc patients with high resistive index, serum levels of endostatin were significantly (p<0.01) higher than in SSc patients with normal resistive index. The serum levels of endostatin significantly increased with progression of nailfold videocapillaroscopy damage (p<0.01) and were significantly (p<0.05) higher in SSc patients with digital ulcers than in SSc patients without digital ulcers. CONCLUSION This is the first study that assess in SSc patients intrarenal hemodynamic parameters and endostatin. In SSc patients, endostatin represents a marker of renal scleroderma-associated vasculopathy.
Collapse
|
42
|
Al Mamun A, Hayashi H, Sakima M, Sato M. Adenosine triphosphate is a critical determinant for VEGFR signal during hypoxia. Am J Physiol Cell Physiol 2016; 311:C985-C995. [PMID: 27834196 DOI: 10.1152/ajpcell.00145.2016] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2016] [Accepted: 10/17/2016] [Indexed: 11/22/2022]
Abstract
Hypoxia induces angiogenesis through the VEGF signaling pathway; however, signal propagation of VEGF in hypoxia is not fully understood. In this study, we examined alterations in VEGF signaling during hypoxia conditions and its determinant in endothelial cells. To analyze VEGF signaling during hypoxia, human umbilical vein endothelial cells (HUVECs) were exposed to 3 h of hypoxia (1% O2) followed by 3 h of reoxygenation or 12 h of hypoxia. Hypoxia induced expression of VEGF mRNA, but it was not associated with an increase in tube formation by HUVECs. During 3 h of hypoxia, VEGF-induced phosphorylation of VEGF receptor-2 (VEGFR-2) and downstream molecules were significantly inhibited without a change in VEGFR-2 expression, but it was completely restored after reoxygenation. VEGF-mediated VEGFR-2 phosphorylation is associated with a reduction in cellular ATP in hypoxia conditions (65.93 ± 8.32% of normoxia, means ± SE, P < 0.01). Interestingly, attenuation of VEGFR-2 phosphorylation was restored by addition of ATP to prepared membranes from cells that underwent 3 h of hypoxia. In contrast to 3 h of hypoxia, exposure of cells to 12 h of hypoxia decreased VEGFR-2 expression and VEGF-mediated VEGFR-2 phosphorylation. The magnitude of VEGFR-2 phosphorylation was not fully restored by addition of ATP to prepared membranes from cells exposed to 12 h of hypoxia. These data indicate that ATP is an important determinant of VEGF signaling in hypoxia and suggest that the activation process of VEGFR-2 was modified by sustained hypoxia. These observations contribute to our understanding of signal alterations in VEGF in endothelial cells during hypoxia.
Collapse
Affiliation(s)
- Abdullah Al Mamun
- Department of Physiology, Aichi Medical University, Nagakute, Aichi, Japan
| | - Hisaki Hayashi
- Department of Physiology, Aichi Medical University, Nagakute, Aichi, Japan
| | - Miho Sakima
- Department of Physiology, Aichi Medical University, Nagakute, Aichi, Japan
| | - Motohiko Sato
- Department of Physiology, Aichi Medical University, Nagakute, Aichi, Japan
| |
Collapse
|
43
|
Saad A, Herrmann SM, Textor SC. Chronic renal ischemia in humans: can cell therapy repair the kidney in occlusive renovascular disease? Physiology (Bethesda) 2016; 30:175-82. [PMID: 25933818 DOI: 10.1152/physiol.00065.2013] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
Occlusive renovascular disease caused by atherosclerotic renal artery stenosis (ARAS) elicits complex biological responses that eventually lead to loss of kidney function. Recent studies indicate a complex interplay of oxidative stress, endothelial dysfunction, and activation of fibrogenic and inflammatory cytokines as a result of atherosclerosis, hypoxia, and renal hypoperfusion in this disorder. Human studies emphasize the limits of the kidney adaptation to reduced blood flow, eventually leading to renal hypoxia with activation of inflammatory and fibrogenic pathways. Several randomized prospective clinical trials show that stent revascularization alone in patients with atherosclerotic renal artery stenosis provides little additional benefit to medical therapy once these processes have developed and solidified. Experimental data now support developing adjunctive cell-based measures to support angiogenesis and anti-inflammatory renal repair mechanisms. These data encourage the study of endothelial progenitor cells and/or mesenchymal stem/stromal cells for the repair of damaged kidney tissue.
Collapse
Affiliation(s)
- Ahmed Saad
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota
| | - Sandra M Herrmann
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota
| | - Stephen C Textor
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota
| |
Collapse
|
44
|
Sequestration of Vascular Endothelial Growth Factor (VEGF) Induces Late Restrictive Lung Disease. PLoS One 2016; 11:e0148323. [PMID: 26863115 PMCID: PMC4749176 DOI: 10.1371/journal.pone.0148323] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2015] [Accepted: 01/15/2016] [Indexed: 11/19/2022] Open
Abstract
Rationale Neonatal respiratory distress syndrome is a restrictive lung disease characterized by surfactant deficiency. Decreased vascular endothelial growth factor (VEGF), which demonstrates important roles in angiogenesis and vasculogenesis, has been implicated in the pathogenesis of restrictive lung diseases. Current animal models investigating VEGF in the etiology and outcomes of RDS require premature delivery, hypoxia, anatomically or temporally limited inhibition, or other supplemental interventions. Consequently, little is known about the isolated effects of chronic VEGF inhibition, started at birth, on subsequent developing lung structure and function. Objectives To determine whether inducible, mesenchyme-specific VEGF inhibition in the neonatal mouse lung results in long-term modulation of AECII and whole lung function. Methods Triple transgenic mice expressing the soluble VEGF receptor sFlt-1 specifically in the mesenchyme (Dermo-1/rtTA/sFlt-1) were generated and compared to littermate controls at 3 months to determine the impact of neonatal downregulation of mesenchymal VEGF expression on lung structure, cell composition and function. Reduced tissue VEGF bioavailability has previously been demonstrated with this model. Measurements and Main Results Triple transgenic mice demonstrated restrictive lung pathology. No differences in gross vascular development or protein levels of vascular endothelial markers was noted, but there was a significant decrease in perivascular smooth muscle and type I collagen. Mutants had decreased expression levels of surfactant protein C and hypoxia inducible factor 1-alpha without a difference in number of type II pneumocytes. Conclusions These data show that mesenchyme-specific inhibition of VEGF in neonatal mice results in late restrictive disease, making this transgenic mouse a novel model for future investigations on the consequences of neonatal RDS and potential interventions.
Collapse
|
45
|
Aplin AC, Nicosia RF. Hypoxia paradoxically inhibits the angiogenic response of isolated vessel explants while inducing overexpression of vascular endothelial growth factor. Angiogenesis 2016; 19:133-46. [PMID: 26748649 DOI: 10.1007/s10456-015-9493-2] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2015] [Accepted: 12/30/2015] [Indexed: 01/08/2023]
Abstract
This study was designed to investigate how changes in O2 levels affected angiogenesis in vascular organ culture. Although hypoxia is a potent inducer of angiogenesis, aortic rings cultured in collagen paradoxically failed to produce an angiogenic response in 1-4 % O2. Additionally, aortic neovessels preformed in atmospheric O2 lost pericytes and regressed at a faster rate than control when exposed to hypoxia. Aortic explants remained viable in hypoxia and produced an angiogenic response when returned to atmospheric O2. Hypoxic aortic rings were unresponsive to VEGF, while increased oxygenation of the system dose-dependently enhanced VEGF-induced angiogenesis. Hypoxia-induced refractoriness to angiogenic stimulation was not restricted to the aorta because similar results were obtained with vena cava explants or isolated endothelial cells. Unlike endothelial cells, aorta-derived mural cells were unaffected by hypoxia. Hypoxia downregulated expression in aortic explants of key signaling molecules including VEGFR2, NRP1 and Prkc-beta while upregulating expression of VEGFR1. Medium conditioned by hypoxic cultures exhibited angiostatic and anti-VEGF activities likely mediated by sVEGFr1. Hypoxia reduced expression of VEGFR1 and VEGFR2 in endothelial cells while upregulating VEGFR1 in macrophages and VEGF in both macrophages and mural cells. Thus, changes in O2 levels profoundly affect the endothelial response to angiogenic stimuli. These results suggest that hypoxia-induced angiogenesis is fine-tuned by complex regulatory mechanisms involving not only production of angiogenic factors including VEGF but also differential regulation of VEGFR expression in different cell types and production of inhibitors of VEGF function such as sVEGFR1.
Collapse
Affiliation(s)
- Alfred C Aplin
- Department of Pathology, University of Washington, Seattle, WA, USA
| | - Roberto F Nicosia
- Department of Pathology, University of Washington, Seattle, WA, USA. .,Pathology and Laboratory Medicine Service (S-113-Lab), VA Puget Sound Health Care System, Seattle, WA, USA.
| |
Collapse
|
46
|
Ulrich F, Carretero-Ortega J, Menéndez J, Narvaez C, Sun B, Lancaster E, Pershad V, Trzaska S, Véliz E, Kamei M, Prendergast A, Kidd KR, Shaw KM, Castranova DA, Pham VN, Lo BD, Martin BL, Raible DW, Weinstein BM, Torres-Vázquez J. Reck enables cerebrovascular development by promoting canonical Wnt signaling. Development 2015; 143:147-59. [PMID: 26657775 DOI: 10.1242/dev.123059] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2015] [Accepted: 11/25/2015] [Indexed: 01/03/2023]
Abstract
The cerebral vasculature provides the massive blood supply that the brain needs to grow and survive. By acquiring distinctive cellular and molecular characteristics it becomes the blood-brain barrier (BBB), a selectively permeable and protective interface between the brain and the peripheral circulation that maintains the extracellular milieu permissive for neuronal activity. Accordingly, there is great interest in uncovering the mechanisms that modulate the formation and differentiation of the brain vasculature. By performing a forward genetic screen in zebrafish we isolated no food for thought (nft (y72)), a recessive late-lethal mutant that lacks most of the intracerebral central arteries (CtAs), but not other brain blood vessels. We found that the cerebral vascularization deficit of nft (y72) mutants is caused by an inactivating lesion in reversion-inducing cysteine-rich protein with Kazal motifs [reck; also known as suppressor of tumorigenicity 15 protein (ST15)], which encodes a membrane-anchored tumor suppressor glycoprotein. Our findings highlight Reck as a novel and pivotal modulator of the canonical Wnt signaling pathway that acts in endothelial cells to enable intracerebral vascularization and proper expression of molecular markers associated with BBB formation. Additional studies with cultured endothelial cells suggest that, in other contexts, Reck impacts vascular biology via the vascular endothelial growth factor (VEGF) cascade. Together, our findings have broad implications for both vascular and cancer biology.
Collapse
Affiliation(s)
- Florian Ulrich
- Dept of Cell Biology, Skirball Institute of Biomolecular Medicine, NYU Langone Medical Center, 540 First Avenue, New York, NY 10016, USA
| | - Jorge Carretero-Ortega
- Dept of Cell Biology, Skirball Institute of Biomolecular Medicine, NYU Langone Medical Center, 540 First Avenue, New York, NY 10016, USA
| | - Javier Menéndez
- Dept of Cell Biology, Skirball Institute of Biomolecular Medicine, NYU Langone Medical Center, 540 First Avenue, New York, NY 10016, USA
| | - Carlos Narvaez
- Dept of Cell Biology, Skirball Institute of Biomolecular Medicine, NYU Langone Medical Center, 540 First Avenue, New York, NY 10016, USA
| | - Belinda Sun
- Dept of Cell Biology, Skirball Institute of Biomolecular Medicine, NYU Langone Medical Center, 540 First Avenue, New York, NY 10016, USA
| | - Eva Lancaster
- Dept of Cell Biology, Skirball Institute of Biomolecular Medicine, NYU Langone Medical Center, 540 First Avenue, New York, NY 10016, USA
| | - Valerie Pershad
- Dept of Cell Biology, Skirball Institute of Biomolecular Medicine, NYU Langone Medical Center, 540 First Avenue, New York, NY 10016, USA
| | - Sean Trzaska
- Dept of Cell Biology, Skirball Institute of Biomolecular Medicine, NYU Langone Medical Center, 540 First Avenue, New York, NY 10016, USA
| | - Evelyn Véliz
- Dept of Cell Biology, Skirball Institute of Biomolecular Medicine, NYU Langone Medical Center, 540 First Avenue, New York, NY 10016, USA
| | - Makoto Kamei
- Program in Genomics of Differentiation, The Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892, USA
| | - Andrew Prendergast
- Department of Biological Structure, University of Washington, Seattle, WA 98195, USA
| | - Kameha R Kidd
- Program in Genomics of Differentiation, The Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892, USA
| | - Kenna M Shaw
- Program in Genomics of Differentiation, The Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892, USA
| | - Daniel A Castranova
- Program in Genomics of Differentiation, The Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892, USA
| | - Van N Pham
- Program in Genomics of Differentiation, The Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892, USA
| | - Brigid D Lo
- Program in Genomics of Differentiation, The Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892, USA
| | | | - David W Raible
- Department of Biological Structure, University of Washington, Seattle, WA 98195, USA
| | - Brant M Weinstein
- Program in Genomics of Differentiation, The Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892, USA
| | - Jesús Torres-Vázquez
- Dept of Cell Biology, Skirball Institute of Biomolecular Medicine, NYU Langone Medical Center, 540 First Avenue, New York, NY 10016, USA
| |
Collapse
|
47
|
Lloyd-Griffith C, McFadden TM, Duffy GP, Unger RE, Kirkpatrick CJ, O’Brien FJ. The pre-vascularisation of a collagen-chondroitin sulphate scaffold using human amniotic fluid-derived stem cells to enhance and stabilise endothelial cell-mediated vessel formation. Acta Biomater 2015; 26:263-73. [PMID: 26300337 DOI: 10.1016/j.actbio.2015.08.030] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2015] [Revised: 08/13/2015] [Accepted: 08/19/2015] [Indexed: 10/23/2022]
Abstract
A major problem in tissue engineering (TE) is graft failure in vivo due to core degradation in in vitro engineered constructs designed to regenerate thick tissues such as bone. The integration of constructs post-implantation relies on the rapid formation of functional vasculature. A recent approach to overcome core degradation focuses on the creation of cell-based, pre-engineered vasculature formed within the TE construct in vitro, prior to implantation in vivo. The primary objective of this study was to investigate whether an amniotic fluid-derived stem cell (AFSC)-human umbilical vein endothelial cell (HUVEC) co-culture could be used to engineer in vitro vasculature in a collagen chondroitin sulphate (CCS) scaffold. The secondary objective was to investigate whether hypoxic conditions (2% O2) could enhance microcapillary-like structure formation by this co-culture. The results of this study demonstrate, for the first time, that the AFSC-HUVEC co-culture was capable of pre-vascularising CCS scaffolds within 7 days and that the AFSCs are capable of behaving as pericytes while interacting with HUVECS to form microcapillary-like structures. However, this microcapillary-like structure formation was reduced in hypoxic conditions. qRT-PCR analysis indicated that an upregulation of VEGFR1 and accompanying decrease of VEGFR2 gene expression may be responsible for the poor response of these microcapillary-like structures to hypoxic conditions. Overall, however, these results demonstrate the potential of this newly developed co-culture system for the formation of pre-engineered vasculature within TE constructs. STATEMENT OF SIGNIFICANCE This article describes the development of an amniotic fluid-derived stem cell (AFSC)-human umbilical vein endothelial cell (HUVEC) co-culture for use in engineering in vitro vasculature in a collagen chondroitin sulphate (CCS) scaffold. The article also describes the effect of hypoxic conditions on the networks of microcapillary-like structures formed by this co-culture. The AFSC-HUVEC co-culture was capable of pre-vascularising CCS scaffolds within 7 days. However, microcapillary-like structure formation was reduced in hypoxic conditions. Overall, these results demonstrate the potential of this newly developed co-culture system for the formation of pre-engineered vasculature within TE constructs. The proangiogenic nature of this co-culture has the potential to both enhance bone regeneration while also overcoming the problem of inadequate vascularisation of grafts commonly seen in the field of tissue engineering.
Collapse
|
48
|
Lee D, Nevo O. 2-Methoxyestradiol regulates VEGFR-2 and sFlt-1 expression in human placenta. Placenta 2015; 36:125-30. [DOI: 10.1016/j.placenta.2014.11.013] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/28/2014] [Revised: 11/11/2014] [Accepted: 11/19/2014] [Indexed: 01/23/2023]
|
49
|
Cuffe JSM, Walton SL, Singh RR, Spiers JG, Bielefeldt-Ohmann H, Wilkinson L, Little MH, Moritz KM. Mid- to late term hypoxia in the mouse alters placental morphology, glucocorticoid regulatory pathways and nutrient transporters in a sex-specific manner. J Physiol 2014; 592:3127-41. [PMID: 24801305 DOI: 10.1113/jphysiol.2014.272856] [Citation(s) in RCA: 89] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Maternal hypoxia is a common perturbation that can disrupt placental and thus fetal development, contributing to neonatal impairments. Recently, evidence has suggested that physiological outcomes are dependent upon the sex of the fetus, with males more susceptible to hypoxic insults than females. This study investigated the effects of maternal hypoxia during mid- to late gestation on fetal growth and placental development and determined if responses were sex specific. CD1 mice were housed under 21% or 12% oxygen from embryonic day (E) 14.5 until tissue collection at E18.5. Fetuses and placentas were weighed before collection for gene and protein expression and morphological analysis. Hypoxia reduced fetal weight in both sexes at E18.5 by 7% but did not affect placental weight. Hypoxia reduced placental mRNA levels of the mineralocorticoid and glucocorticoid receptors and reduced the gene and protein expression of the glucocorticoid metabolizing enzyme HSD11B2. However, placentas of female fetuses responded differently to maternal hypoxia than did placentas of male fetuses. Notably, morphology was significantly altered in placentas from hypoxic female fetuses, with a reduction in placental labyrinth blood spaces. In addition mRNA expression of Glut1, Igf2 and Igf1r were reduced in placentas of female fetuses only. In summary, maternal hypoxia altered placental formation in a sex specific manner through mechanisms involving placental vascular development, growth factor and nutrient transporter expression and placental glucocorticoid signalling. This study provides insight into how sex differences in offspring disease development may be due to sex specific placental adaptations to maternal insults.
Collapse
Affiliation(s)
- J S M Cuffe
- School of Biomedical Sciences, The University of Queensland, St Lucia, Queensland, Australia
| | - S L Walton
- School of Biomedical Sciences, The University of Queensland, St Lucia, Queensland, Australia
| | - R R Singh
- School of Biomedical Sciences, The University of Queensland, St Lucia, Queensland, Australia
| | - J G Spiers
- School of Biomedical Sciences, The University of Queensland, St Lucia, Queensland, Australia
| | - H Bielefeldt-Ohmann
- School of Veterinary Science, The University of Queensland, Gatton, Queensland, Australia
| | - L Wilkinson
- Institute for Molecular Bioscience, The University of Queensland, St Lucia, Queensland, Australia
| | - M H Little
- Institute for Molecular Bioscience, The University of Queensland, St Lucia, Queensland, Australia
| | - K M Moritz
- School of Biomedical Sciences, The University of Queensland, St Lucia, Queensland, Australia
| |
Collapse
|
50
|
Alderson HV, Ritchie JP, Kalra PA. Revascularization as a treatment to improve renal function. Int J Nephrol Renovasc Dis 2014; 7:89-99. [PMID: 24600242 PMCID: PMC3933706 DOI: 10.2147/ijnrd.s35633] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
An aging atherosclerosis-prone population has led to an increase in the prevalence of atherosclerotic renovascular disease (ARVD). Medical management of this disease, as with other atherosclerotic conditions, has improved over the past decade. Despite the widespread availability of endovascular revascularization procedures, there is inconsistent evidence of benefit in ARVD and no clear consensus of opinion as to the best way to select suitable patients for revascularization. Several published randomized controlled trials have attempted to provide clearer evidence for best practice in ARVD, but they have done so with varying clarity and success. In this review, we provide an overview of ARVD and its effect on renal function. We present the currently available evidence for best practice in the management of patients with ARVD with a particular focus on revascularization as a treatment to improve renal function. We provide a brief overview of the evidence for revascularization in other causes of renal artery stenosis.
Collapse
Affiliation(s)
- Helen V Alderson
- The University of Manchester, Manchester Academic Health Sciences Centre, Vascular Research Group, Department of Renal Medicine, Salford Royal NHS Foundation Trust, Salford, UK
| | - James P Ritchie
- The University of Manchester, Manchester Academic Health Sciences Centre, Vascular Research Group, Department of Renal Medicine, Salford Royal NHS Foundation Trust, Salford, UK
| | - Philip A Kalra
- The University of Manchester, Manchester Academic Health Sciences Centre, Vascular Research Group, Department of Renal Medicine, Salford Royal NHS Foundation Trust, Salford, UK
| |
Collapse
|