1
|
Nys N, Khatib AM, Siegfried G. Apela promotes blood vessel regeneration and remodeling in zebrafish. Sci Rep 2024; 14:3718. [PMID: 38355946 PMCID: PMC10867005 DOI: 10.1038/s41598-023-50677-1] [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: 08/28/2023] [Accepted: 12/22/2023] [Indexed: 02/16/2024] Open
Abstract
In contrast to adult mammals, zebrafish display a high capacity to heal injuries and repair damage to various organs. One of the earliest responses to injury in adult zebrafish is revascularization, followed by tissue morphogenesis. Tissue vascularization entails the formation of a blood vessel plexus that remodels into arteries and veins. The mechanisms that coordinate these processes during vessel regeneration are poorly understood. Hence, investigating and identifying the factors that promote revascularization and vessel remodeling have great therapeutic potential. Here, we revealed that fin vessel remodeling critically depends on Apela peptide. We found that Apela selectively accumulated in newly formed zebrafish fin tissue and vessels. The temporal expression of Apela, Apln, and their receptor Aplnr is different during the regenerative process. While morpholino-mediated knockdown of Apela (Mo-Apela) prevented vessel remodeling, exogenous Apela peptide mediated plexus repression and the development of arteries in regenerated fins. In contrast, Apela enhanced subintestinal venous plexus formation (SIVP). The use of sunitinib completely inhibited vascular plexus formation in zebrafish, which was not prevented by exogenous application. Furthermore, Apela regulates the expression of vessel remolding-related genes including VWF, IGFPB3, ESM1, VEGFR2, Apln, and Aplnr, thereby linking Apela to the vascular plexus factor network as generated by the STRING online database. Together, our findings reveal a new role for Apela in vessel regeneration and remodeling in fin zebrafish and provide a framework for further understanding the cellular and molecular mechanisms involved in vessel regeneration.
Collapse
Affiliation(s)
- Nicolas Nys
- RYTME, Bordeaux Institute of Oncology (BRIC)-UMR1312 Inserm, University of Bordeaux, B2 Ouest, Allée Geoffroy St Hilaire CS50023, 33615, Pessac, France
| | - Abdel-Majid Khatib
- RYTME, Bordeaux Institute of Oncology (BRIC)-UMR1312 Inserm, University of Bordeaux, B2 Ouest, Allée Geoffroy St Hilaire CS50023, 33615, Pessac, France.
- ZebraFish, Research and Technology, B2 Ouest, Allée Geoffroy St Hilaire CS50023, 33615, Pessac, France.
- Bergonié Institute, Bordeaux, France.
| | - Geraldine Siegfried
- RYTME, Bordeaux Institute of Oncology (BRIC)-UMR1312 Inserm, University of Bordeaux, B2 Ouest, Allée Geoffroy St Hilaire CS50023, 33615, Pessac, France.
- ZebraFish, Research and Technology, B2 Ouest, Allée Geoffroy St Hilaire CS50023, 33615, Pessac, France.
| |
Collapse
|
2
|
Xu Y, Miller CP, Xue J, Zheng Y, Warren EH, Tykodi SS, Akilesh S. Single cell atlas of kidney cancer endothelial cells reveals distinct expression profiles and phenotypes. RESEARCH SQUARE 2023:rs.3.rs-3558517. [PMID: 37986984 PMCID: PMC10659545 DOI: 10.21203/rs.3.rs-3558517/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/22/2023]
Abstract
Background Tumor endothelial cells (TECs) represent the primary interface between the tumor microenvironment and circulating immune cells, however their phenotypes are incompletely understood in highly vascularized clear cell renal cell carcinoma (ccRCC). Methods We purified tumor and matched normal endothelial cells (NECs) from ccRCC specimens and performed single-cell RNA-sequencing to create a reference-quality atlas available as a searchable web resource for gene expression patterns. We established paired primary TECs and NECs cultures for ex vivo functional testing. Results TECs from multiple donors shared a common phenotype with increased expression of pathways related to extracellular matrix regulation, cell-cell communication, and insulin-like growth factor signaling that was conserved in comparison to hepatocellular carcinoma associated TECs, suggesting convergent TEC phenotypes between unrelated tumors. Cultured TECs stably maintained a core program of differentially regulated genes, were inherently resistant to apoptosis after vascular endothelial growth factor removal and displayed increased adhesiveness to subsets of immune cells including regulatory T-cells. Conclusions Our studies delineate unique functional and phenotypic properties of TECs, which may provide insights into their interactions with available and emerging therapies. Functional phenotypes of cultured TECs suggest potential mechanisms of resistance to both antiangiogenic and immune-based therapies.
Collapse
|
3
|
Maitra P, Prema S, Narendran V, Shah PK. Safety and efficacy of intravitreal anti vascular endothelial growth factor for severe posterior retinopathy of prematurity with flat fibrovascular proliferation. World J Clin Pediatr 2023; 12:220-229. [PMID: 37753496 PMCID: PMC10518743 DOI: 10.5409/wjcp.v12.i4.220] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Revised: 06/26/2023] [Accepted: 07/07/2023] [Indexed: 09/06/2023] Open
Abstract
BACKGROUND Intravitreal anti-vascular endothelial growth factor (IVA) injection is known to cause contraction of fibrovascular proliferation (FVP), when present in severe retinopathy of prematurity (ROP). AIM To assess the structural outcomes of IVA injection in the treatment of severe posterior ROP with significant FVP. METHODS It was a retrospective study in which 36 eyes of 18 preterm babies who developed > 4 clock hours of FVP in zone I or posterior zone II, were treated with either intravitreal 0.625 mg bevacizumab or intravitreal 0.2 mg of ranibizumab. Favorable structural outcome included resolution of plus disease and FVP without the development of tractional retinal detachment. Secondary outcome measure included either full retinal maturation at follow-up or development of recurrent disease requiring additional treatment. Adverse outcomes included progression to retinal detachment. RESULTS The mean gestational age of the 18 preterm babies was 30 wk (range 27-36), and mean birth weight was 1319 g (range 650-1980 g). Mean post-menstrual age (PMA) at the time of primary treatment was 35.5 wk (range 31-41 wk). All eyes showed regression of plus disease and FVP. 5 eyes of 3 babies showed reactivation of disease and were treated with repeat IVA (n = 2 eyes) or peripheral laser photocoagulation (n = 3 eyes) respectively. 16 out of 36 (44%) reached retinal vascular maturation at final follow up at 5 years. CONCLUSION There was good resolution of severe posterior ROP with FVP with IVA, with retinal maturity of 44% at 5 year follow-up and a reactivation rate of 13.8%. When the IVA injection is given prior to 37 wk PMA, while disease is in phase 2, it is less likely to cause contracture of pre-existing FVP.
Collapse
Affiliation(s)
- Puja Maitra
- Pediatric Retina and Ocular Oncology, Aravind Eye Hospital and Postgraduate Institute of Ophthalmology, Coimbatore 641014, Tamil Nadu, India
| | - Subramaniam Prema
- Pediatric Retina and Ocular Oncology, Aravind Eye Hospital and Postgraduate Institute of Ophthalmology, Coimbatore 641014, Tamil Nadu, India
| | - Venkatapathy Narendran
- Pediatric Retina and Ocular Oncology, Aravind Eye Hospital and Postgraduate Institute of Ophthalmology, Coimbatore 641014, Tamil Nadu, India
| | - Parag K Shah
- Pediatric Retina and Ocular Oncology, Aravind Eye Hospital and Postgraduate Institute of Ophthalmology, Coimbatore 641014, Tamil Nadu, India
| |
Collapse
|
4
|
Prasad M, Ingolfsland EC, Christiansen SP. Modifiable Risk Factors and Preventative Strategies for Severe Retinopathy of Prematurity. Life (Basel) 2023; 13:life13051075. [PMID: 37240719 DOI: 10.3390/life13051075] [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: 03/06/2023] [Revised: 04/13/2023] [Accepted: 04/13/2023] [Indexed: 05/28/2023] Open
Abstract
Severe ROP is characterized by the development of retinal fibrovascular proliferation that may progress to retinal detachment. The purpose of this report is to review five of the most common and well-studied perinatal and neonatal modifiable risk factors for the development of severe ROP. Hyperoxemia, hypoxia, and associated prolonged respiratory support are linked to the development of severe ROP. While there is a well-established association between clinical maternal chorioamnionitis and severe ROP, there is greater variability between histologic chorioamnionitis and severe ROP. Neonatal sepsis, including both bacterial and fungal subtypes, are independent predictors of severe ROP in preterm infants. Although there is limited evidence related to platelet transfusions, the risk of severe ROP increases with the number and volume of red blood cell transfusions. Poor postnatal weight gain within the first six weeks of life is also strongly tied to the development of severe ROP. We also discuss preventative strategies that may reduce the risk of severe ROP. Limited evidence-based studies exist regarding the protective effects of caffeine, human milk, and vitamins A and E.
Collapse
Affiliation(s)
- Minali Prasad
- Boston University Chobanian & Avedisian School of Medicine, Boston, MA 02118, USA
| | - Ellen C Ingolfsland
- Department of Pediatrics, Division of Neonatology, University of Minnesota, Minneapolis, MN 55455, USA
| | - Stephen P Christiansen
- Departments of Ophthalmology and Pediatrics, Boston University Chobanian & Avedisian School of Medicine, Boston Medical Center, Boston, MA 02118, USA
| |
Collapse
|
5
|
Fevereiro-Martins M, Marques-Neves C, Guimarães H, Bicho M. Retinopathy of prematurity: A review of pathophysiology and signaling pathways. Surv Ophthalmol 2023; 68:175-210. [PMID: 36427559 DOI: 10.1016/j.survophthal.2022.11.007] [Citation(s) in RCA: 26] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Revised: 11/15/2022] [Accepted: 11/18/2022] [Indexed: 11/25/2022]
Abstract
Retinopathy of prematurity (ROP) is a vasoproliferative disorder of the retina and a leading cause of visual impairment and childhood blindness worldwide. The disease is characterized by an early stage of retinal microvascular degeneration, followed by neovascularization that can lead to subsequent retinal detachment and permanent visual loss. Several factors play a key role during the different pathological stages of the disease. Oxidative and nitrosative stress and inflammatory processes are important contributors to the early stage of ROP. Nitric oxide synthase and arginase play important roles in ischemia/reperfusion-induced neurovascular degeneration. Destructive neovascularization is driven by mediators of the hypoxia-inducible factor pathway, such as vascular endothelial growth factor and metabolic factors (succinate). The extracellular matrix is involved in hypoxia-induced retinal neovascularization. Vasorepulsive molecules (semaphorin 3A) intervene preventing the revascularization of the avascular zone. This review focuses on current concepts about signaling pathways and their mediators, involved in the pathogenesis of ROP, highlighting new potentially preventive and therapeutic modalities. A better understanding of the intricate molecular mechanisms underlying the pathogenesis of ROP should allow the development of more effective and targeted therapeutic agents to reduce aberrant vasoproliferation and facilitate physiological retinal vascular development.
Collapse
Affiliation(s)
- Mariza Fevereiro-Martins
- Laboratório de Genética and Grupo Ecogenética e Saúde Humana, Instituto de Saúde Ambiental, Faculdade de Medicina, Universidade de Lisboa, Portugal; Instituto de Investigação Científica Bento da Rocha Cabral, Lisboa, Portugal; Departamento de Oftalmologia, Hospital Cuf Descobertas, Lisboa, Portugal.
| | - Carlos Marques-Neves
- Centro de Estudos das Ci.¼ncias da Visão, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal; Grupo Ecogenética e Saúde Humana, Instituto de Saúde Ambiental, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal.
| | - Hercília Guimarães
- Departamento de Ginecologia-Obstetrícia e Pediatria, Faculdade de Medicina, Universidade do Porto, Porto, Portugal.
| | - Manuel Bicho
- Laboratório de Genética and Grupo Ecogenética e Saúde Humana, Instituto de Saúde Ambiental, Faculdade de Medicina, Universidade de Lisboa, Portugal; Instituto de Investigação Científica Bento da Rocha Cabral, Lisboa, Portugal.
| |
Collapse
|
6
|
Systemic Cytokines in Retinopathy of Prematurity. J Pers Med 2023; 13:jpm13020291. [PMID: 36836525 PMCID: PMC9966226 DOI: 10.3390/jpm13020291] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Revised: 01/30/2023] [Accepted: 01/30/2023] [Indexed: 02/09/2023] Open
Abstract
Retinopathy of prematurity (ROP), a vasoproliferative vitreoretinal disorder, is the leading cause of childhood blindness worldwide. Although angiogenic pathways have been the main focus, cytokine-mediated inflammation is also involved in ROP etiology. Herein, we illustrate the characteristics and actions of all cytokines involved in ROP pathogenesis. The two-phase (vaso-obliteration followed by vasoproliferation) theory outlines the evaluation of cytokines in a time-dependent manner. Levels of cytokines may even differ between the blood and the vitreous. Data from animal models of oxygen-induced retinopathy are also valuable. Although conventional cryotherapy and laser photocoagulation are well established and anti-vascular endothelial growth factor agents are available, less destructive novel therapeutics that can precisely target the signaling pathways are required. Linking the cytokines involved in ROP to other maternal and neonatal diseases and conditions provides insights into the management of ROP. Suppressing disordered retinal angiogenesis via the modulation of hypoxia-inducible factor, supplementation of insulin-like growth factor (IGF)-1/IGF-binding protein 3 complex, erythropoietin, and its derivatives, polyunsaturated fatty acids, and inhibition of secretogranin III have attracted the attention of researchers. Recently, gut microbiota modulation, non-coding RNAs, and gene therapies have shown promise in regulating ROP. These emerging therapeutics can be used to treat preterm infants with ROP.
Collapse
|
7
|
Xin J, He Y, Guo K, Yang D. Expression of the neuroprotective factors BDNF, CNTF, and FGF-2 in normal and oxygen induced retinopathy. Front Neurosci 2022; 16:971952. [PMID: 36532277 PMCID: PMC9755753 DOI: 10.3389/fnins.2022.971952] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Accepted: 11/18/2022] [Indexed: 11/30/2023] Open
Abstract
INTRODUCTION Oxygen-induced retinopathy is a type of retinal pathological neovascularization (NV) disease that leads to vision loss and translates to a significant societal cost. Anti-vascular endothelial growth factor (VEGF) and anti-inflammatory treatments have been widely used in the clinic, but the results have not been entirely satisfactory. It is necessary to explore other treatments for Ischemic retinal diseases. METHODS The oxygen-induced retinopathy (OIR) model was induced from P7 to P12 as described. Histology evaluation (HE) and retina flat mounts were checked at P17 to confirm the establishment of the OIR model. Retinal ganglion cell (RGC) degeneration was checked by transmission electron microscopy at P17 to confirm the neurological damage caused by OIR. Western blot analysis was performed at P12, P15, and P17 to study the expression of brain-derived neurotrophic factor (BDNF), ciliary neurotrophic factor (CNTF), and fibroblast growth factor 2 (FGF-2) in normal and OIR mice. Comparative analysis of the expressions of BDNF, CNTF, and FGF-2 in normal and OIR mice was performed. RESULTS There were many retinal NV and non-perfusion areas in OIR P17. RGCs were degenerated at OIR P17. The expressions of BDNF, CNTF, and FGF-2 gradually increased from P12 to P17 in normal mice and were much higher in OIR mice. The expression curves of BDNF, CNTF, and FGF-2 in the OIR model were inconsistent and did not correlate with each other. DISCUSSION This study provides evidence for changes in BDNF, CNTF, and FGF-2 in Oxygen-induced retinopathy.
Collapse
Affiliation(s)
| | | | - Kai Guo
- Department of Ophthalmology, The Affiliated Hospital of Inner Mongolia Medical University, Hohhot, China
| | - Dayong Yang
- Department of Ophthalmology, The Affiliated Hospital of Inner Mongolia Medical University, Hohhot, China
| |
Collapse
|
8
|
Lyu T, Yang X, Zhao C, Wang L, Zhou S, Shi L, Dong Y, Dou H, Zhang H. Comparative transcriptomics of high-altitude Vulpes and their low-altitude relatives. Front Ecol Evol 2022. [DOI: 10.3389/fevo.2022.999411] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
The harsh environment of Qinghai-Tibet Plateau (QTP) imposes strong selective stresses (e.g., hypoxia, high UV-radiation, and extreme temperature) to the native species, which have driven striking phenotypic and genetic adaptations. Although the mechanisms of high-altitude adaptation have been explored for many plateau species, how the phylogenetic background contributes to genetic adaption to high-altitude of Vulpes is largely unknown. In this study, we sequenced transcriptomic data across multiple tissues of two high-altitude Vulpes (Vulpes vulpes montana and Vulpes ferrilata) and their low-altitude relatives (Vulpes corsac and Vulpes lagopus) to search the genetic and gene expression changes caused by high-altitude environment. The results indicated that the positive selection genes (PSGs) identified by both high-altitude Vulpes are related to angiogenesis, suggesting that angiogenesis may be the result of convergent evolution of Vulpes in the face of hypoxic selection pressure. In addition, more PSGs were detected in V. ferrilata than in V. v. montana, which may be related to the longer adaptation time of V. ferrilata to plateau environment and thus more genetic changes. Besides, more PSGs associated with high-altitude adaptation were identified in V. ferrilata compared with V. v. montana, indicating that the longer the adaptation time to the high-altitude environment, the more genetic alterations of the species. Furthermore, the result of expression profiles revealed a tissue-specific pattern between Vulpes. We also observed that differential expressed genes in the high-altitude group exhibited species-specific expression patterns, revealed a convergent expression pattern of Vulpes in high-altitude environment. In general, our research provides a valuable transcriptomic resource for further studies, and expands our understanding of high-altitude adaptation within a phylogenetic context.
Collapse
|
9
|
Qin K, Lei J, Yang J. The Differentiation of Pluripotent Stem Cells towards Endothelial Progenitor Cells - Potential Application in Pulmonary Arterial Hypertension. Int J Stem Cells 2021; 15:122-135. [PMID: 34711697 PMCID: PMC9148829 DOI: 10.15283/ijsc21044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2021] [Revised: 08/26/2021] [Accepted: 09/02/2021] [Indexed: 11/22/2022] Open
Abstract
Background and Objectives Endothelial progenitor cells (EPCs) and endothelial cells (ECs) have been applied in the clinic to treat pulmonary arterial hypertension (PAH), a disease characterized by disordered pulmonary vasculature. However, the lack of sufficient transplantable cells before the deterioration of disease condition is a current limitation to apply cell therapy in patients. It is necessary to differentiate pluripotent stem cells (PSCs) into EPCs and identify their characteristics. Methods and Results Comparing previously reported methods of human PSCs-derived ECs, we optimized a highly efficient differentiation protocol to obtain cells that match the phenotype of isolated EPCs from healthy donors. The protocol is compatible with chemically defined medium (CDM), it could produce a large number of clinically applicable cells with low cost. Moreover, we also found PSCs-derived EPCs express CD133, have some characteristics of mesenchymal stem cells and are capable of homing to repair blood vessels in zebrafish xenograft assays. In addition, we further revealed that IPAH PSCs-derived EPCs have higher expression of proliferation-related genes and lower expression of immune-related genes than normal EPCs and PSCs-derived EPCs through microarray analysis. Conclusions In conclusion, we optimized a highly efficient differentiation protocol to obtain PSCs-derived EPCs with the phenotypic and molecular characteristics of EPCs from healthy donors which distinguished them from EPCs from PAH.
Collapse
Affiliation(s)
- Kezhou Qin
- Department of Cell Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Jia Lei
- Department of Physiology, and Department of Cardiology of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Jun Yang
- Department of Cell Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China.,Department of Physiology, and Department of Cardiology of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
10
|
Zhang K, Li F, Yan B, Xiao DJ, Wang YS, Liu H. Comparison of the Cytokine Profile in Mesenchymal Stem Cells from Human Adipose, Umbilical Cord, and Placental Tissues. Cell Reprogram 2021; 23:336-348. [PMID: 34677101 DOI: 10.1089/cell.2021.0043] [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] [Indexed: 12/29/2022] Open
Abstract
Human mesenchymal stem cells (MSCs) can be isolated from various tissues. However, the cytokine profile in different MSC types remains unclear. In this study, MSCs were extracted from adipose, umbilical cord, and placental tissues. The surface marker expression, multilineage differentiation potential, and cytokine secretion of these cells were compared. The isolated MSCs exhibited similar morphology and surface marker expression. However, they differed with regard to their differentiation potential. Adipose-MSCs (A-MSCs) exhibited a higher potential for adipogenesis and osteogenic differentiation compared with umbilical cord-MSCs (UC-MSCs) and placental-MSCs (P-MSCs). The expression levels of 80 cytokines were detected, and the data demonstrated that the three MSC types abundantly secreted insulin-like growth factor-binding protein (IGFBP)-4, IGFBP-3, tissue inhibitor of metalloproteinase (TIMP)-1, TIMP-2, IGFBP-6, monocyte chemoattractant protein-1, and granulocyte colony-stimulating factor. However, the expression levels of vascular endothelial growth factor, tumor necrosis factor alpha, interleukin (IL)-6 receptor, and IL-13 in A-MSCs were higher compared with those of UC-MSCs and P-MSCs. Moreover, the expression levels of intercellular adhesion molecule-1 and growth differentiation factor 15 were lower in A-MSCs. Kyoto Encyclopedia of Genes and Genomes analysis indicated that the "adipocytokine" and the "PI3K/Akt pathways" were enriched in A-MSCs. Taken together, the results demonstrated that MSCs from different sources exhibited differences in the secretion of specific factors. A-MSCs were associated with the expression of several proangiogenic factors and may be an improved source for angiogenesis and tissue regeneration.
Collapse
Affiliation(s)
- Kun Zhang
- Cell Therapy Center, Jinan Central Hospital, Jinan, P.R. China.,Jinan Central Hospital, Cheeloo College of Medicine, Shandong University, Jinan, P.R. China.,Shandong Research Center of Transplantation and Tissue, Jinan, P.R. China
| | - Fang Li
- Cell Therapy Center, Jinan Central Hospital, Jinan, P.R. China.,Jinan Central Hospital, Cheeloo College of Medicine, Shandong University, Jinan, P.R. China.,Shandong Research Center of Transplantation and Tissue, Jinan, P.R. China
| | - Bing Yan
- Department of Gastrointestinal Surgery, Jinan Central Hospital, Cheeloo College of Medicine, Shandong University, Jinan, P.R. China
| | - Dong-Jie Xiao
- Cell Therapy Center, Jinan Central Hospital, Jinan, P.R. China.,Jinan Central Hospital, Cheeloo College of Medicine, Shandong University, Jinan, P.R. China
| | - Yun-Shan Wang
- Cell Therapy Center, Jinan Central Hospital, Jinan, P.R. China.,Jinan Central Hospital, Cheeloo College of Medicine, Shandong University, Jinan, P.R. China
| | - Hua Liu
- Cell Therapy Center, Jinan Central Hospital, Jinan, P.R. China.,Jinan Central Hospital, Cheeloo College of Medicine, Shandong University, Jinan, P.R. China.,Shandong Research Center of Transplantation and Tissue, Jinan, P.R. China
| |
Collapse
|
11
|
Kurmann L, Okoniewski M, Dubey RK. Transcryptomic Analysis of Human Brain -Microvascular Endothelial Cell Driven Changes in -Vascular Pericytes. Cells 2021; 10:cells10071784. [PMID: 34359953 PMCID: PMC8304094 DOI: 10.3390/cells10071784] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Revised: 06/30/2021] [Accepted: 07/09/2021] [Indexed: 12/12/2022] Open
Abstract
Many pathological conditions of the brain are associated with structural abnormalities within the neurovascular system and linked to pericyte (PC) loss and/or dysfunction. Since crosstalk between endothelial cells (ECs) and PCs greatly impacts the function of the blood–brain barrier (BBB), effects of PCs on endothelial integrity and function have been investigated extensively. However, the impact of ECs on the function and activity of PCs remains largely unknown. Hence, using co-cultures of human brain vascular PCs with human cerebral microvascular ECs on opposite sides of porous Transwell inserts which facilitates direct EC–PC contact and improves EC barrier function, we analyzed EC-driven transcriptomic changes in PCs using microarrays and changes in cytokines/chemokines using proteome arrays. Gene expression analysis (GEA) in PCs co-cultured with ECs versus PCs cultured alone showed significant upregulation of 1′334 genes and downregulation of 964 genes. GEA in co-cultured PCs revealed increased expression of five prominent PC markers as well as soluble factors, such as transforming growth factor beta, fibroblast growth factor, angiopoietin 1, brain-derived neurotrophic factor, all of which are involved in EC–PC crosstalk and BBB induction. Pathway enrichment analysis of modulated genes showed a strong impact on many inflammatory and extracellular matrix (ECM) pathways including interferon and interleukin signaling, TGF-β and interleukin-1 regulation of ECM, as well as on the mRNA processing pathway. Interestingly, while co-culture induced the mRNA expression of many chemokines and cytokines, including several CCL- and CXC-motif ligands and interleukins, we observed a decreased expression of the same inflammatory mediators on the protein level. Importantly, in PCs, ECs significantly induced interferon associated proteins (IFIT1, IFI44L, IF127, IFIT3, IFI6, IFI44) with anti-viral actions; downregulated prostaglandin E receptor 2 (prevent COX-2 mediated BBB damage); upregulated fibulin-3 and connective tissue growth factor essential for BBB integrity; and multiple ECMs (collagens and integrins) that inhibit cell migration. Our findings suggest that via direct contact, ECs prime PCs to induce molecules to promote BBB integrity and cell survival during infection and inflammatory insult. Taken together, we provide first evidence that interaction with ECs though porous membranes induces major changes in the transcriptomic and proteomic profile of PCs. ECs influence genes involved in diverse aspects of PC function including PC maturation, cell survival, anti-viral defense, blood flow regulation, immuno-modulation and ECM deposition.
Collapse
Affiliation(s)
- Lisa Kurmann
- Department of Reproductive Endocrinology, University Hospital Zurich, 8952 Schlieren, Switzerland;
| | | | - Raghvendra K. Dubey
- Department of Reproductive Endocrinology, University Hospital Zurich, 8952 Schlieren, Switzerland;
- Department of Pharmacology & Chemical Biology, University of Pittsburgh, Pittsburgh, PA 15219, USA
- Correspondence:
| |
Collapse
|
12
|
Dai C, Webster KA, Bhatt A, Tian H, Su G, Li W. Concurrent Physiological and Pathological Angiogenesis in Retinopathy of Prematurity and Emerging Therapies. Int J Mol Sci 2021; 22:4809. [PMID: 34062733 PMCID: PMC8124946 DOI: 10.3390/ijms22094809] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Revised: 04/27/2021] [Accepted: 04/29/2021] [Indexed: 12/11/2022] Open
Abstract
Retinopathy of prematurity (ROP) is an ocular vascular disease affecting premature infants, characterized by pathological retinal neovascularization (RNV), dilated and tortuous retinal blood vessels, and retinal or vitreous hemorrhages that may lead to retinal detachment, vision impairment and blindness. Compared with other neovascular diseases, ROP is unique because of ongoing and concurrent physiological and pathological angiogenesis in the developing retina. While the disease is currently treated by laser or cryotherapy, anti-vascular endothelial growth factor (VEGF) agents have been extensively investigated but are not approved in the U.S. because of safety concerns that they negatively interfere with physiological angiogenesis of the developing retina. An ideal therapeutic strategy would selectively inhibit pathological but not physiological angiogenesis. Our group recently described a novel strategy that selectively and safely alleviates pathological RNV in animal models of ROP by targeting secretogranin III (Scg3), a disease-restricted angiogenic factor. The preclinical profile of anti-Scg3 therapy presents a high potential for next-generation disease-targeted anti-angiogenic therapy for the ROP indication. This review focuses on retinal vessel development in neonates, the pathogenesis of ROP and its underlying molecular mechanisms, including different animal models, and provides a summary of current and emerging therapies.
Collapse
Affiliation(s)
- Chang Dai
- Department of Ophthalmology, Baylor College of Medicine, Houston, TX 77030, USA; (C.D.); (K.A.W.); (A.B.)
- Department of Ophthalmology, The Second Hospital of Jilin University, Changchun 130041, China
| | - Keith A. Webster
- Department of Ophthalmology, Baylor College of Medicine, Houston, TX 77030, USA; (C.D.); (K.A.W.); (A.B.)
- Department of Pharmacology, University of Miami School of Medicine, Miami, FL 33136, USA
- Everglades Biopharma, LLC, Houston, TX 77030, USA;
| | - Amit Bhatt
- Department of Ophthalmology, Baylor College of Medicine, Houston, TX 77030, USA; (C.D.); (K.A.W.); (A.B.)
- Texas Children Hospital, Baylor College of Medicine, Houston, TX 77030, USA
| | - Hong Tian
- Everglades Biopharma, LLC, Houston, TX 77030, USA;
| | - Guanfang Su
- Department of Ophthalmology, The Second Hospital of Jilin University, Changchun 130041, China
| | - Wei Li
- Department of Ophthalmology, Baylor College of Medicine, Houston, TX 77030, USA; (C.D.); (K.A.W.); (A.B.)
| |
Collapse
|
13
|
Tisch N, Ruiz de Almodóvar C. Contribution of cell death signaling to blood vessel formation. Cell Mol Life Sci 2021; 78:3247-3264. [PMID: 33783563 PMCID: PMC8038986 DOI: 10.1007/s00018-020-03738-x] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Revised: 12/06/2020] [Accepted: 12/08/2020] [Indexed: 02/07/2023]
Abstract
The formation of new blood vessels is driven by proliferation of endothelial cells (ECs), elongation of maturing vessel sprouts and ultimately vessel remodeling to create a hierarchically structured vascular system. Vessel regression is an essential process to remove redundant vessel branches in order to adapt the final vessel density to the demands of the surrounding tissue. How exactly vessel regression occurs and whether and to which extent cell death contributes to this process has been in the focus of several studies within the last decade. On top, recent findings challenge our simplistic view of the cell death signaling machinery as a sole executer of cellular demise, as emerging evidences suggest that some of the classic cell death regulators even promote blood vessel formation. This review summarizes our current knowledge on the role of the cell death signaling machinery with a focus on the apoptosis and necroptosis signaling pathways during blood vessel formation in development and pathology.
Collapse
Affiliation(s)
- Nathalie Tisch
- Department of Vascular Dysfunction, European Center for Angioscience (ECAS), Faculty of Medicine Mannheim, University of Heidelberg, Mannheim, Germany
| | - Carmen Ruiz de Almodóvar
- Department of Vascular Dysfunction, European Center for Angioscience (ECAS), Faculty of Medicine Mannheim, University of Heidelberg, Mannheim, Germany.
| |
Collapse
|
14
|
Young J, Bell S, Qian Y, Hyman C, Berryman DE. Mouse models of growth hormone insensitivity. Rev Endocr Metab Disord 2021; 22:17-29. [PMID: 33037595 PMCID: PMC7979446 DOI: 10.1007/s11154-020-09600-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 10/01/2020] [Indexed: 11/28/2022]
Abstract
Growth hormone (GH) induces pleiotropic effects on growth and metabolism via binding and subsequent activation of the growth hormone receptor (GHR) and its downstream signaling pathways. Growth hormone insensitivity (GHI) describes a group of disorders in which there is resistance to the action of GH and resultant insulin-like growth factor I (IGF-I) deficiency. GHI is commonly due to genetic disorders of the GH receptor causing GH receptor deficiency (e.g. Laron Syndrome (LS)), decreased activation of GHR, or defects in post-receptor signaling molecules. Genetically altered mouse lines have been invaluable to better understand the physiological impact of GHI due to the ability to do invasive and longitudinal measures of metabolism, growth, and health on a whole animal or in individual tissues/cells. In the current review, the phenotype of mouse lines with GHI will be reviewed. Mouse lines to be discussed include: 1) GHR-/- mice with a gene disruption in the GHR that results in no functional GHR throughout life, also referred to as the Laron mouse, 2) mice with temporal loss of GHR (aGHRKO) starting at 6 weeks of age, 3) mice transgenic for a GHR antagonist (GHA mice), 4) mice with GHI in select tissues or cells generated via Cre-lox or related technology, and 5) assorted mice with defects in post-receptor signaling molecules. Collectively, these mouse lines have revealed an intriguing role of GH action in health, disease, and aging.
Collapse
Affiliation(s)
- Jonathan Young
- Department of Biomedical Sciences, Ohio University Heritage College of Osteopathic Medicine, Athens, OH, 45701, USA
- Edison Biotechnology Institute, Konneker Research Labs, Ohio University, Athens, OH, USA
| | - Stephen Bell
- Department of Biomedical Sciences, Ohio University Heritage College of Osteopathic Medicine, Athens, OH, 45701, USA
- Edison Biotechnology Institute, Konneker Research Labs, Ohio University, Athens, OH, USA
| | - Yanrong Qian
- Edison Biotechnology Institute, Konneker Research Labs, Ohio University, Athens, OH, USA
| | - Caroline Hyman
- Department of Biomedical Sciences, Ohio University Heritage College of Osteopathic Medicine, Athens, OH, 45701, USA
| | - Darlene E Berryman
- Department of Biomedical Sciences, Ohio University Heritage College of Osteopathic Medicine, Athens, OH, 45701, USA.
- Edison Biotechnology Institute, Konneker Research Labs, Ohio University, Athens, OH, USA.
| |
Collapse
|
15
|
Arima M, Fujii Y, Sonoda KH. Translational Research in Retinopathy of Prematurity: From Bedside to Bench and Back Again. J Clin Med 2021; 10:331. [PMID: 33477419 PMCID: PMC7830975 DOI: 10.3390/jcm10020331] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Revised: 01/09/2021] [Accepted: 01/15/2021] [Indexed: 12/11/2022] Open
Abstract
Retinopathy of prematurity (ROP), a vascular proliferative disease affecting preterm infants, is a leading cause of childhood blindness. Various studies have investigated the pathogenesis of ROP. Clinical experience indicates that oxygen levels are strongly correlated with ROP development, which led to the development of oxygen-induced retinopathy (OIR) as an animal model of ROP. OIR has been used extensively to investigate the molecular mechanisms underlying ROP and to evaluate the efficacy of new drug candidates. Large clinical trials have demonstrated the efficacy of anti-vascular endothelial growth factor (VEGF) agents to treat ROP, and anti-VEGF therapy is presently becoming the first-line treatment worldwide. Anti-VEGF therapy has advantages over conventional treatments, including being minimally invasive with a low risk of refractive error. However, long-term safety concerns and the risk of late recurrence limit this treatment. There is an unmet medical need for novel ROP therapies, which need to be addressed by safe and minimally invasive therapies. The recent progress in biotechnology has contributed greatly to translational research. In this review, we outline how basic ROP research has evolved with clinical experience and the subsequent emergence of new drugs. We discuss previous and ongoing trials and present the candidate molecules expected to become novel targets.
Collapse
Affiliation(s)
- Mitsuru Arima
- Department of Ophthalmology, Graduate School of Medical Sciences, Kyushu University, Fukuoka 8128582, Japan; (Y.F.); (K.-H.S.)
- Center for Clinical and Translational Research, Kyushu University Hospital, 3-1-1 Maidashi, Higashi-ku, Fukuoka 8128582, Japan
| | - Yuya Fujii
- Department of Ophthalmology, Graduate School of Medical Sciences, Kyushu University, Fukuoka 8128582, Japan; (Y.F.); (K.-H.S.)
| | - Koh-Hei Sonoda
- Department of Ophthalmology, Graduate School of Medical Sciences, Kyushu University, Fukuoka 8128582, Japan; (Y.F.); (K.-H.S.)
| |
Collapse
|
16
|
Zhang WB, Aleksic S, Gao T, Weiss EF, Demetriou E, Verghese J, Holtzer R, Barzilai N, Milman S. Insulin-like Growth Factor-1 and IGF Binding Proteins Predict All-Cause Mortality and Morbidity in Older Adults. Cells 2020; 9:cells9061368. [PMID: 32492897 PMCID: PMC7349399 DOI: 10.3390/cells9061368] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2020] [Revised: 05/27/2020] [Accepted: 05/28/2020] [Indexed: 01/02/2023] Open
Abstract
While the growth hormone/insulin-like growth factor-1 (GH/IGF-1) pathway plays essential roles in growth and development, diminished signaling via this pathway in model organisms extends lifespan and health-span. In humans, circulating IGF-1 and IGF-binding proteins 3 and 1 (IGFBP-3 and 1), surrogate measures of GH/IGF-1 system activity, have not been consistently associated with morbidity and mortality. In a prospective cohort of independently-living older adults (n = 840, mean age 76.1 ± 6.8 years, 54.5% female, median follow-up 6.9 years), we evaluated the age- and sex-adjusted hazards for all-cause mortality and incident age-related diseases, including cardiovascular disease, diabetes, cancer, and multiple-domain cognitive impairment (MDCI), as predicted by baseline total serum IGF-1, IGF-1/IGFBP-3 molar ratio, IGFBP-3, and IGFBP-1 levels. All-cause mortality was positively associated with IGF-1/IGFBP-3 molar ratio (HR 1.28, 95% CI 1.05–1.57) and negatively with IGFBP-3 (HR 0.82, 95% CI 0.680–0.998). High serum IGF-1 predicted greater risk for MDCI (HR 1.56, 95% CI 1.08–2.26) and composite incident morbidity (HR 1.242, 95% CI 1.004–1.538), whereas high IGFBP-1 predicted lower risk for diabetes (HR 0.50, 95% CI 0.29–0.88). In conclusion, higher IGF-1 levels and bioavailability predicted mortality and morbidity risk, supporting the hypothesis that diminished GH/IGF-1 signaling may contribute to human longevity and health-span.
Collapse
Affiliation(s)
- William B. Zhang
- Department of Medicine, Division of Endocrinology, Institute for Aging Research, Albert Einstein College of Medicine, Bronx, NY 10461, USA; (W.B.Z.); (S.A.); (T.G.); (N.B.)
| | - Sandra Aleksic
- Department of Medicine, Division of Endocrinology, Institute for Aging Research, Albert Einstein College of Medicine, Bronx, NY 10461, USA; (W.B.Z.); (S.A.); (T.G.); (N.B.)
| | - Tina Gao
- Department of Medicine, Division of Endocrinology, Institute for Aging Research, Albert Einstein College of Medicine, Bronx, NY 10461, USA; (W.B.Z.); (S.A.); (T.G.); (N.B.)
| | - Erica F. Weiss
- Department of Neurology, Albert Einstein College of Medicine, Bronx, NY 10461, USA; (E.F.W.); (J.V.); (R.H.)
| | - Eleni Demetriou
- Ferkauf Graduate School of Psychology, Yeshiva University, New York, NY 10033, USA;
| | - Joe Verghese
- Department of Neurology, Albert Einstein College of Medicine, Bronx, NY 10461, USA; (E.F.W.); (J.V.); (R.H.)
- Department of Medicine, Division of Geriatrics, Institute for Aging Research, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Roee Holtzer
- Department of Neurology, Albert Einstein College of Medicine, Bronx, NY 10461, USA; (E.F.W.); (J.V.); (R.H.)
- Ferkauf Graduate School of Psychology, Yeshiva University, New York, NY 10033, USA;
| | - Nir Barzilai
- Department of Medicine, Division of Endocrinology, Institute for Aging Research, Albert Einstein College of Medicine, Bronx, NY 10461, USA; (W.B.Z.); (S.A.); (T.G.); (N.B.)
- Department of Genetics, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Sofiya Milman
- Department of Medicine, Division of Endocrinology, Institute for Aging Research, Albert Einstein College of Medicine, Bronx, NY 10461, USA; (W.B.Z.); (S.A.); (T.G.); (N.B.)
- Department of Genetics, Albert Einstein College of Medicine, Bronx, NY 10461, USA
- Correspondence:
| |
Collapse
|
17
|
Cai Q, Dozmorov M, Oh Y. IGFBP-3/IGFBP-3 Receptor System as an Anti-Tumor and Anti-Metastatic Signaling in Cancer. Cells 2020; 9:cells9051261. [PMID: 32443727 PMCID: PMC7290346 DOI: 10.3390/cells9051261] [Citation(s) in RCA: 66] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Revised: 05/15/2020] [Accepted: 05/18/2020] [Indexed: 12/11/2022] Open
Abstract
Insulin-like growth factor binding protein-3 (IGFBP-3) is a p53 tumor suppressor-regulated protein and a major carrier for IGFs in circulation. Among six high-affinity IGFBPs, which are IGFBP-1 through 6, IGFBP-3 is the most extensively investigated IGFBP species with respect to its IGF/IGF-I receptor (IGF-IR)-independent biological actions beyond its endocrine/paracrine/autocrine role in modulating IGF action in cancer. Disruption of IGFBP-3 at transcriptional and post-translational levels has been implicated in the pathophysiology of many different types of cancer including breast, prostate, and lung cancer. Over the past two decades, a wealth of evidence has revealed both tumor suppressing and tumor promoting effects of IGF/IGF-IR-independent actions of IGFBP-3 depending upon cell types, post-translational modifications, and assay methods. However, IGFBP-3′s anti-tumor function has been well accepted due to identification of functional IGFBP-3-interacting proteins, putative receptors, or crosstalk with other signaling cascades. This review mainly focuses on transmembrane protein 219 (TMEM219), which represents a novel IGFBP-3 receptor mediating antitumor effect of IGFBP-3. Furthermore, this review delineates the potential underlying mechanisms involved and the subsequent biological significance, emphasizing the clinical significance of the IGFBP-3/TMEM219 axis in assessing both the diagnosis and the prognosis of cancer as well as the therapeutic potential of TMEM219 agonists for cancer treatment.
Collapse
Affiliation(s)
- Qing Cai
- Department of Pathology, Medical College of Virginia Campus, Virginia Commonwealth University, Richmond, VA 23298, USA; (Q.C.); (M.D.)
| | - Mikhail Dozmorov
- Department of Pathology, Medical College of Virginia Campus, Virginia Commonwealth University, Richmond, VA 23298, USA; (Q.C.); (M.D.)
- Department of Biostatistics, Massey Cancer Center, Virginia Commonwealth University, Richmond, VA 23298, USA
| | - Youngman Oh
- Department of Pathology, Medical College of Virginia Campus, Virginia Commonwealth University, Richmond, VA 23298, USA; (Q.C.); (M.D.)
- Correspondence: ; Tel.: +1-804-827-1324
| |
Collapse
|
18
|
Tiemann J, Wagner T, Vanakker OM, van Gils M, Cabrera JLB, Ibold B, Faust I, Knabbe C, Hendig D. Cellular and Molecular Biomarkers Indicate Premature Aging in Pseudoxanthoma Elasticum Patients. Aging Dis 2020; 11:536-546. [PMID: 32489700 PMCID: PMC7220280 DOI: 10.14336/ad.2019.0610] [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: 04/17/2019] [Accepted: 06/05/2019] [Indexed: 01/09/2023] Open
Abstract
The molecular processes of aging are very heterogenic and not fully understood. Studies on rare progeria syndromes, which display an accelerated progression of physiological aging, can help to get a better understanding. Pseudoxanthoma elasticum (PXE) caused by mutations in the ATP-binding cassette sub-family C member 6 (ABCC6) gene shares some molecular characteristics with such premature aging diseases. Thus, this is the first study trying to broaden the knowledge of aging processes in PXE patients. In this study, we investigated aging associated biomarkers in primary human dermal fibroblasts and sera from PXE patients compared to healthy controls. Determination of serum concentrations of the aging biomarkers eotaxin-1 (CCL11), growth differentiation factor 11 (GDF11) and insulin-like growth factor 1 (IGF1) showed no significant differences between PXE patients and healthy controls. Insulin-like growth factor binding protein 3 (IGFBP3) showed a significant increase in serum concentrations of PXE patients older than 45 years compared to the appropriate control group. Tissue specific gene expression of GDF11 and IGFBP3 were significantly decreased in fibroblasts from PXE patients compared to normal human dermal fibroblasts (NHDF). IGFBP3 protein concentration in supernatants of fibroblasts from PXE patients were decreased compared to NHDF but did not reach statistical significance due to potential gender specific variations. The minor changes in concentration of circulating aging biomarkers in sera of PXE patients and the significant aberrant tissue specific expression seen for selected factors in PXE fibroblasts, suggests a link between ABCC6 deficiency and accelerated aging processes in affected peripheral tissues of PXE patients.
Collapse
Affiliation(s)
- Janina Tiemann
- 1Institut für Laboratoriums- und Transfusionsmedizin, Herz- und Diabeteszentrum Nordrhein-Westfalen, Universitätsklinik der Ruhr-Universität Bochum, Bad Oeynhausen, Germany
| | - Thomas Wagner
- 1Institut für Laboratoriums- und Transfusionsmedizin, Herz- und Diabeteszentrum Nordrhein-Westfalen, Universitätsklinik der Ruhr-Universität Bochum, Bad Oeynhausen, Germany
| | | | - Matthias van Gils
- 2Center for Medical Genetics, Ghent University Hospital, Ghent, Belgium
| | - José-Luis Bueno Cabrera
- 3Haematology Department, Hospital Universitario Puerta de Hierro-Majadahonda, Majadahonda, Spain
| | - Bettina Ibold
- 1Institut für Laboratoriums- und Transfusionsmedizin, Herz- und Diabeteszentrum Nordrhein-Westfalen, Universitätsklinik der Ruhr-Universität Bochum, Bad Oeynhausen, Germany
| | - Isabel Faust
- 1Institut für Laboratoriums- und Transfusionsmedizin, Herz- und Diabeteszentrum Nordrhein-Westfalen, Universitätsklinik der Ruhr-Universität Bochum, Bad Oeynhausen, Germany
| | - Cornelius Knabbe
- 1Institut für Laboratoriums- und Transfusionsmedizin, Herz- und Diabeteszentrum Nordrhein-Westfalen, Universitätsklinik der Ruhr-Universität Bochum, Bad Oeynhausen, Germany
| | - Doris Hendig
- 1Institut für Laboratoriums- und Transfusionsmedizin, Herz- und Diabeteszentrum Nordrhein-Westfalen, Universitätsklinik der Ruhr-Universität Bochum, Bad Oeynhausen, Germany
| |
Collapse
|
19
|
Kumar R, Mani AM, Singh NK, Rao GN. PKCθ-JunB axis via upregulation of VEGFR3 expression mediates hypoxia-induced pathological retinal neovascularization. Cell Death Dis 2020; 11:325. [PMID: 32382040 PMCID: PMC7206019 DOI: 10.1038/s41419-020-2522-0] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2019] [Revised: 04/17/2020] [Accepted: 04/20/2020] [Indexed: 12/30/2022]
Abstract
Pathological retinal neovascularization is the most common cause of vision loss. PKCθ has been shown to play a role in type 2 diabetes, which is linked to retinal neovascularization. Based on these clues, we have studied the role of PKCθ and its downstream target genes JunB and VEGFR3 in retinal neovascularization using global and tissue-specific knockout mouse models along with molecular biological approaches. Here, we show that vascular endothelial growth factor A (VEGFA) induces PKCθ phosphorylation in human retinal microvascular endothelial cells (HRMVECs) and downregulation of its levels attenuates VEGFA-induced HRMVECs migration, sprouting and tube formation. Furthermore, the whole body deletion of PKCθ or EC-specific deletion of its target gene JunB inhibited hypoxia-induced retinal EC proliferation, tip cell formation and neovascularization. VEGFA also induced VEGFR3 expression via JunB downstream to PKCθ in the regulation of HRMVEC migration, sprouting, and tube formation in vitro and OIR-induced retinal EC proliferation, tip cell formation and neovascularization in vivo. In addition, VEGFA-induced VEGFR3 expression requires VEGFR2 activation upstream to PKCθ-JunB axis both in vitro and in vivo. Depletion of VEGFR2 or VEGFR3 levels attenuated VEGFA-induced HRMVEC migration, sprouting and tube formation in vitro and retinal neovascularization in vivo and it appears that these events were dependent on STAT3 activation. Furthermore, the observations using soluble VEGFR3 indicate that VEGFR3 mediates its effects on retinal neovascularization in a ligand dependent and independent manner downstream to VEGFR2. Together, these observations suggest that PKCθ-dependent JunB-mediated VEGFR3 expression targeting STAT3 activation is required for VEGFA/VEGFR2-induced retinal neovascularization.
Collapse
Affiliation(s)
- Raj Kumar
- Department of Physiology, University of Tennessee Health Science Center, Memphis, TN, 38163, USA
| | - Arul M Mani
- Department of Physiology, University of Tennessee Health Science Center, Memphis, TN, 38163, USA
| | - Nikhlesh K Singh
- Department of Physiology, University of Tennessee Health Science Center, Memphis, TN, 38163, USA
| | - Gadiparthi N Rao
- Department of Physiology, University of Tennessee Health Science Center, Memphis, TN, 38163, USA.
| |
Collapse
|
20
|
Carroll L, Owen LA. Current evidence and outcomes for retinopathy of prematurity prevention: insight into novel maternal and placental contributions. EXPLORATION OF MEDICINE 2020; 1:4-26. [PMID: 32342063 PMCID: PMC7185238 DOI: 10.37349/emed.2020.00002] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2019] [Accepted: 11/27/2019] [Indexed: 12/11/2022] Open
Abstract
Retinopathy of prematurity (ROP) is a blinding morbidity of preterm infants, which represents a significant clinical problem, accounting for up to 40% of all childhood blindness. ROP displays a range of severity, though even mild disease may result in life-long visual impairment. This is complicated by the fact that our current treatments have significant ocular and potentially systemic effects. Therefore, disease prevention is desperately needed to mitigate the life-long deleterious effects of ROP for preterm infants. Although ROP demonstrates a delayed onset of retinal disease following preterm birth, representing a potential window for prevention, we have been unable to sufficiently alter the natural disease course and meaningfully prevent ROP. Prevention therapeutics requires knowledge of early ROP molecular changes and risk, occurring prior to clinical retinal disease. While we still have an incomplete understanding of these disease mechanisms, emerging data integrating contributions of maternal/placental pathobiology with ROP are poised to inform novel approaches to prevention. Herein, we review the molecular basis for current prevention strategies and the clinical outcomes of these interventions. We also discuss how insights into early ROP pathophysiology may be gained by a better understanding of maternal and placental factors playing a role in preterm birth.
Collapse
Affiliation(s)
- Lara Carroll
- Department of Ophthalmology and Visual Sciences, University of Utah, Salt Lake City, UT 4132, USA
| | - Leah A. Owen
- Department of Ophthalmology and Visual Sciences, University of Utah, Salt Lake City, UT 4132, USA
| |
Collapse
|
21
|
Pirouzpanah S, Asemani S, Shayanfar A, Baradaran B, Montazeri V. The effects of Berberis vulgaris consumption on plasma levels of IGF-1, IGFBPs, PPAR-γ and the expression of angiogenic genes in women with benign breast disease: a randomized controlled clinical trial. BMC COMPLEMENTARY AND ALTERNATIVE MEDICINE 2019; 19:324. [PMID: 31752829 PMCID: PMC6868871 DOI: 10.1186/s12906-019-2715-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/09/2019] [Accepted: 10/14/2019] [Indexed: 01/10/2023]
Abstract
BACKGROUND The present study was designed to investigate the effects of Berberis vulgaris (BV) juice consumption on plasma levels of insulin-like growth factor (IGF-1), IGF-binding proteins (IGFBPs), and the expression of PPAR-γ, VEGF and HIF in women with benign breast disease. METHODS This parallel design randomized, double-blind controlled clinical trial was conducted on 85 eligible patients diagnosed with benign breast disease. They were assigned randomly into either BV juice group (n = 44, BV juice: 480 ml/day) or placebo group (n = 41, BV placebo juice: 480 ml/day) for 8 weeks intervention. Participants, caregivers and those who assessed laboratory analyses were blinded to the assignments. Plasma levels of biomarkers were measured at baseline and after 8 weeks by ELISA. Quantitative real-time PCR was used to measure the fold change in the expression of each interested gene. RESULTS The compliance of participants was 95.2% and 40 available subjects analyzed in each group at last. Relative treatment (RT) effects for BV juice caused 16% fall in IGF-1 concentration and 37% reduction in the ratio of IGF-1/1GFBP1. Absolute treatment effect expressed 111 ng/ml increased mean differences of IGFBP-3 between BV group and placebo. Plasma level of PPAR-γ increased in both groups but it was not significant. Fold changes in the expressions of PPAR-γ, VEGF and HIF showed down-regulation in the intervention group compared to placebos (P < 0.05). CONCLUSIONS The BV juice intervention over 8 weeks was accompanied by acceptable efficacy and decreased plasma IGF-1, and IGF-1/IGFBP-1 ratio partly could be assigned to enhanced IGFBP-1 level in women with BBD. The intervention caused reductions in the expression levels of PPAR, VEGF, and HIF which are remarkable genomic changes to potentially prevent breast tumorigenesis. TRIAL REGISTRATION IRCT2012110511335N2. Registered 10 July 2013 (retrospectively registered).
Collapse
|
22
|
Liu L, Jiang Y, Steinle JJ. Glycyrrhizin protects IGFBP-3 knockout mice from retinal damage. Cytokine 2019; 125:154856. [PMID: 31526985 DOI: 10.1016/j.cyto.2019.154856] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2019] [Revised: 09/09/2019] [Accepted: 09/10/2019] [Indexed: 11/28/2022]
Abstract
We previously reported that insulin-like growth factor binding protein 3 (IGFBP-3) knockout (KO) mice have neuronal and vascular damage to the retina. We also reported that glycyrrhizin, a high mobility growth factor binding protein 1 (HMGB1) inhibitor, is protective to the diabetic retina. In this study, we investigated whether glycyrrhizin could reduce neuronal and vascular damage in the IGFBP-3 KO mouse retina. We used measurements of retinal thickness, cell number in the ganglion cell layer, degenerate capillaries, reactive oxygen species (ROS) and protein levels of HMGB1, tumor necrosis factor alpha (TNFα), interleukin-1-beta (IL-1β) and sirtuin 1 (SIRT1) to determine whether glycyrrhizin could protect the retina. Data show that glycyrrhizin in the drinking water was effective in reducing neuronal damage at 2 months and vascular damage at 6 months. Glycyrrhizin reduced ROS levels at 6 months, and reduced levels of HMGB1, TNFα, and IL-1β at both 2 and 6 months. Taken together, the data suggest that glycyrrhizin is protective to the retina of IGFBP-3 KO mice through anti-inflammatory mechanisms.
Collapse
Affiliation(s)
- Li Liu
- Department of Ophthalmology, Visual, and Anatomical Sciences, Wayne State University School of Medicine, Detroit, MI 48201, USA
| | - Youde Jiang
- Department of Ophthalmology, Visual, and Anatomical Sciences, Wayne State University School of Medicine, Detroit, MI 48201, USA
| | - Jena J Steinle
- Department of Ophthalmology, Visual, and Anatomical Sciences, Wayne State University School of Medicine, Detroit, MI 48201, USA.
| |
Collapse
|
23
|
Bai R, Cui Z, Ma Y, Wu Y, Wang N, Huang L, Yao Q, Sun J. The NF-κB-modulated miR-19a-3p enhances malignancy of human ovarian cancer cells through inhibition of IGFBP-3 expression. Mol Carcinog 2019; 58:2254-2265. [PMID: 31513316 DOI: 10.1002/mc.23113] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2019] [Revised: 08/14/2019] [Accepted: 08/29/2019] [Indexed: 12/15/2022]
Abstract
Ovarian cancer is the most lethal gynecologic malignancy due to the lack of symptoms until advanced stages, and new diagnosis and treatment strategy is in urgent need. In this study, we found higher expression of miR-19a-3p in ovarian cancer tissues compared with that in the adjacent normal tissues. By chromatin immunoprecipitation (ChIP) and electrophoretic mobility shift assay (EMSA) analysis, we showed that nuclear factor-kappaB (NF-κB) binds to the promoter of miR-19a-3p, leading to reduced expression in ovarian cancer cells. Further study indicated that miR-19a-3p inhibits the expression of insulin-like growth factor binding protein-3 (IGFBP-3), resulting in enhanced growth and migration of ovarian cancer cells in vitro and tumor growth in vivo. These results showed that miR-19a-3p enhances the oncogenesis of ovarian cancer through inhibition of IGFBP-3 expression, and which can be inhibited by NF-κB, suggesting an NF-κB/miR-19a-3p/IGFBP-3 pathway in the oncogenesis of ovarian cancer, which expands our understanding of ovarian cancer and they may contribute to the development of new diagnosis and treatment of ovarian cancer.
Collapse
Affiliation(s)
- Ru Bai
- Department of Pathogen Biology and Immunoly, School of Basic Medical Sciences, Ningxia Medical University, Yinchuan, China
| | - Zhenhua Cui
- Department of Pathogen Biology and Immunoly, School of Basic Medical Sciences, Ningxia Medical University, Yinchuan, China
| | - Yongjing Ma
- Department of Gynecological Tumors Surgery, General Hospital of Ningxia Medical University, Yinchuan, China
| | - Yang Wu
- Department of Pathogen Biology and Immunoly, School of Pharmacy, Ningxia Medical University, Yinchuan, China
| | - Ningping Wang
- Department of Pathogen Biology and Immunoly, School of Basic Medical Sciences, Ningxia Medical University, Yinchuan, China
| | - Ling Huang
- Department of Pathogen Biology and Immunoly, School of Basic Medical Sciences, Ningxia Medical University, Yinchuan, China
| | - Qing Yao
- Department of Pathogen Biology and Immunoly, School of Basic Medical Sciences, Ningxia Medical University, Yinchuan, China
| | - Jianmin Sun
- Department of Pathogen Biology and Immunoly, School of Basic Medical Sciences, Ningxia Medical University, Yinchuan, China.,Division of Translational Cancer Research, Department of Laboratory Medicine, Lund Stem Cell Center, Lund University, Lund, Sweden
| |
Collapse
|
24
|
Renes JS, van Doorn J, Hokken-Koelega ACS. Current Insights into the Role of the Growth Hormone-Insulin-Like Growth Factor System in Short Children Born Small for Gestational Age. Horm Res Paediatr 2019; 92:15-27. [PMID: 31509834 PMCID: PMC6979433 DOI: 10.1159/000502739] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/22/2019] [Accepted: 08/14/2019] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND The reason for the insufficient catch-up growth seen in 10% of children born small for gestational age (SGA) is poorly understood. Disturbances in the growth hormone (GH) - insulin-like growth factor (IGF) axis might underlie this failure to show sufficient catch-up growth. CONCLUSION This review summarizes insights gained in the molecular and (epi) genetic mechanisms of the GH-IGF axis in short children born SGA. The most notable anomalies of the IGF system are the lowered IGF-I levels in both cord blood and the placenta, and the increased expression of IGF-binding proteins (IGFBP)-1 and IGFBP-2, which inhibit IGF-I, in the placenta of SGA neonates. These observations suggest a decreased bioactivity of IGF-I in utero. IGF-I levels remain reduced in SGA children with short stature, as well as IGFBP-3 and acid-labile subunit levels. Proteolysis of IGFBP-3 appears to be increased.
Collapse
Affiliation(s)
- Judith S Renes
- Department of Paediatrics, Subdivision of Endocrinology, Erasmus University Medical Centre, Sophia Children's Hospital, Rotterdam, The Netherlands,
| | - Jaap van Doorn
- Department of Genetics, Section of Metabolic Diagnostics, University Medical Centre Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Anita C S Hokken-Koelega
- Department of Paediatrics, Subdivision of Endocrinology, Erasmus University Medical Centre, Sophia Children's Hospital, Rotterdam, The Netherlands
- Dutch Growth Research Foundation, Rotterdam, The Netherlands
| |
Collapse
|
25
|
Tsang JKW, Liu J, Lo ACY. Vascular and Neuronal Protection in the Developing Retina: Potential Therapeutic Targets for Retinopathy of Prematurity. Int J Mol Sci 2019; 20:E4321. [PMID: 31484463 PMCID: PMC6747312 DOI: 10.3390/ijms20174321] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2019] [Revised: 08/21/2019] [Accepted: 08/29/2019] [Indexed: 12/13/2022] Open
Abstract
Retinopathy of prematurity (ROP) is a common retinal disease in preterm babies. To prolong the lives of preterm babies, high oxygen is provided to mimic the oxygen level in the intrauterine environment for postnatal organ development. However, hyperoxia-hypoxia induced pathological events occur when babies return to room air, leading to ROP with neuronal degeneration and vascular abnormality that affects retinal functions. With advances in neonatal intensive care, it is no longer uncommon for increased survival of very-low-birth-weight preterm infants, which, therefore, increased the incidence of ROP. ROP is now a major cause of preventable childhood blindness worldwide. Current proven treatment for ROP is limited to invasive retinal ablation, inherently destructive to the retina. The lack of pharmacological treatment for ROP creates a great need for effective and safe therapies in these developing infants. Therefore, it is essential to identify potential therapeutic agents that may have positive ROP outcomes, especially in preserving retinal functions. This review gives an overview of various agents in their efficacy in reducing retinal damages in cell culture tests, animal experiments and clinical studies. New perspectives along the neuroprotective pathways in the developing retina are also reviewed.
Collapse
Affiliation(s)
- Jessica K W Tsang
- Department of Ophthalmology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Jin Liu
- Department of Ophthalmology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Amy C Y Lo
- Department of Ophthalmology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China.
| |
Collapse
|
26
|
Sheikhrezaee M, Alizadeh MR, Abediankenari S. The tear VEGF and IGFBP3 in healthy and diabetic retinopathy. Int J Diabetes Dev Ctries 2019. [DOI: 10.1007/s13410-019-00761-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
|
27
|
Abstract
Introduction Retinopathy of prematurity (ROP) is a leading cause of childhood blindness worldwide. Areas covered Recent methods to identify and manage treatment-warranted vascularly active ROP are recognized and being compared to standard care by laser treatment in prospective large-scale clinical studies. Pharmacologic anti-angiogenic (anti-VEGF) treatment has changed the natural history of vascularly active ROP by reducing stage 3 intravitreal neovascularization and extending physiologic retinal vascularization in many infants. Tractional retinal detachments in stage 4 ROP after treatment with anti-VEGF agents show additional fibrovascular complexity compared to eyes treated with laser only. We review current management and outcomes for vascularly active and fibrovascular retinal detachment in ROP (stages 3, 4, 5 ROP), highlighting the evidence from recent clinical studies. Included are technical details important in surgery for retinal detachment in ROP. Literature searches were employed through PubMed. Expert opinion Methods in pediatric imaging, safer pharmacologic treatments, and surgical techniques continue to advance to improve future ROP outcomes.
Collapse
Affiliation(s)
- Eric D Hansen
- John A. Moran Eye Center, University of Utah, Salt Lake City, USA
| | | |
Collapse
|
28
|
Slater T, Haywood NJ, Matthews C, Cheema H, Wheatcroft SB. Insulin-like growth factor binding proteins and angiogenesis: from cancer to cardiovascular disease. Cytokine Growth Factor Rev 2019; 46:28-35. [PMID: 30954375 DOI: 10.1016/j.cytogfr.2019.03.005] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2019] [Accepted: 03/15/2019] [Indexed: 12/22/2022]
Abstract
Angiogenesis is a tightly regulated activity that is vital during embryonic development and for normal physiological repair processes and reproduction in healthy adults. Pathological angiogenesis is a driving force behind a variety of diseases including cancer and retinopathies, and inhibition of angiogenesis is a therapeutic option that has been the subject of much research, with several inhibitory agents now available for medical therapy. Conversely, therapeutic angiogenesis has been mooted as having significant potential in the treatment of ischemic conditions such as angina pectoris and peripheral arterial disease, but so far there has been less translation from lab to bedside. The insulin-like growth factor binding proteins (IGFBP) are a family of seven proteins essential for the binding and transport of the insulin-like growth factors (IGF). It is being increasingly recognised that IGFBPs have a significant role beyond simply modulating IGF activity, with evidence of both IGF dependent and independent actions through a variety of mechanisms. Moreover, the action of the IGFBPs can be stimulatory or inhibitory depending on the cell type and environment. Specifically the IGFBPs have been heavily implicated in angiogenesis, both pathological and physiological, and they have significant promise as targeted cell therapy agents for both pathological angiogenesis inhibition and therapeutic angiogenesis following ischemic injury. In this short review we will explore the current understanding of the individual impact of each IGFBP on angiogenesis, and the pathways through which these effects occur.
Collapse
Affiliation(s)
- Thomas Slater
- Leeds Institute of Cardiovascular & Metabolic Medicine, School of Medicine, University of Leeds, United Kingdom
| | - Natalie J Haywood
- Leeds Institute of Cardiovascular & Metabolic Medicine, School of Medicine, University of Leeds, United Kingdom
| | - Connor Matthews
- Leeds Institute of Cardiovascular & Metabolic Medicine, School of Medicine, University of Leeds, United Kingdom
| | - Harneet Cheema
- Leeds Institute of Cardiovascular & Metabolic Medicine, School of Medicine, University of Leeds, United Kingdom
| | - Stephen B Wheatcroft
- Leeds Institute of Cardiovascular & Metabolic Medicine, School of Medicine, University of Leeds, United Kingdom.
| |
Collapse
|
29
|
The Effect of Serum Igf-1,Igfbp-3 And Erythrocyte Transfusıons on Development of Mıld Retınopathy of Prematurıty. JOURNAL OF CONTEMPORARY MEDICINE 2019. [DOI: 10.16899/gopctd.535602] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
|
30
|
Meng X, Sun B, Xiao Z. Comparison in transcriptome and cytokine profiles of mesenchymal stem cells from human umbilical cord and cord blood. Gene 2019; 696:10-20. [PMID: 30769140 DOI: 10.1016/j.gene.2019.02.017] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2018] [Revised: 01/23/2019] [Accepted: 02/08/2019] [Indexed: 12/14/2022]
Abstract
Human umbilical cord (UC) and cord blood (CB) provide attractive sources of mesenchymal stem cells (MSCs) for cell therapy. Both UCMSCs and CBMSCs have been demonstrated to play prominent roles in clinical therapy. However, little is known about their functional differences in clinical application. Our transcriptome analysis uncovered high activity of insulin secretion related signaling pathways for CBMSCs and cell adhesion related signaling pathways for UCMSCs. Expression of a large number of immune related signaling pathways also showed the difference in both cells, implying their distinct immune modulatory functions. As the therapeutic effects of MSCs mainly dependent on the cytokines and growth factors produced by transplanted MSCs, we further compared the cytokine profiles of UCMSCs and CBMSCs using antibody array. By evaluating the expression of 106 cytokines, we found both MSCs abundantly secreted TSP-1, TSG-14, TIMP-1, IL-8, IL-6, CXCL1, GIF and IGFBP3. However, the expression of CCL2 in UCMSCs showed significantly higher than CBMSCs. IGFBP1 and IGFBP2 were secreted by CBMSCs with higher abundance than UCMSCs. Overall, these results suggest that UCMSCs and CBMSCs preserve different functional potentials, which have to be carefully considered before clinical treatment.
Collapse
Affiliation(s)
- Xianhui Meng
- State key laboratory of bioelectronics, School of biological science & medical engineering, Southeast University, Nanjing, 210096, China
| | - Bo Sun
- State key laboratory of bioelectronics, School of biological science & medical engineering, Southeast University, Nanjing, 210096, China.
| | - Zhongdang Xiao
- State key laboratory of bioelectronics, School of biological science & medical engineering, Southeast University, Nanjing, 210096, China.
| |
Collapse
|
31
|
Jafari E, Gheysarzadeh A, Mahnam K, Shahmohammadi R, Ansari A, Bakhtyari H, Mofid MR. In silico interaction of insulin-like growth factor binding protein 3 with insulin-like growth factor 1. Res Pharm Sci 2018; 13:332-342. [PMID: 30065766 PMCID: PMC6040160 DOI: 10.4103/1735-5362.235160] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Insulin-like growth factor binding protein-3 (IGFBP-3) is a vital protein exist in circulation which interacts with high affinity to insulin-like growth factor (IGFs) altering their activities. Therefore, the interaction between IGFs and IGFBP-3 has a key role altering large spectrum of activities such as cell cycle progression, proliferation and apoptosis. Despite decades of research, the crystal structure of IGFBP-3 has not been identified possibly due to some technical challenge in its crystallizing. The three-dimensional (3D) structure of IGFBP-3 was predicted using homology modeling, Phyre2, and molecular dynamic. Its interaction with IGF-1 was also identified by HADDOCK software. IGFBP-3 has the most identity with other IGFBPs in N and C-domain; however, its linker domain has lower identity. Our data predicted that IGF-1 structurally interacts with N-domain and linker domain of IGFBP-3. Some conserved residues of IGFBP-3 such as Glu33, Arg36, Gly39, Arg60, Arg66, Asn109, and Ile146 interacts with Glu3, Asp12, Phe16, Gly19, Asp20, Arg21, and Glu58 of IGF-1. In addition, our data predict that the linker domain has a loop structure which covers post translational modification and interacts with IGF-1. The phosphorylation of Ser111 in linker domain, which previously has been shown to induce apoptosis make a repulsive force interrupting this interaction to IGF-1, which enables IGFBP-3 to induce apoptosis. The present study suggests that the linker domain has a key role in recognition of IGFBP-3 with IGF-1.
Collapse
Affiliation(s)
- Elham Jafari
- Department of Medicinal Chemistry, Bioinformatics Research Center, School of Pharmacy and Pharmaceutical Sciences, Isfahan University of Medical Sciences, Isfahan, I.R. Iran
| | - Ali Gheysarzadeh
- Department of Biochemistry and Bioinformatics Research Center, School of Pharmacy and Pharmaceutical Sciences, Isfahan University of Medical Sciences, Isfahan, I.R. Iran
| | - Karim Mahnam
- Biology Department, Faculty of Sciences, Shahrekord University, Shahrekord, I.R. Iran
| | | | - Amir Ansari
- Department of Biochemistry and Bioinformatics Research Center, School of Pharmacy and Pharmaceutical Sciences, Isfahan University of Medical Sciences, Isfahan, I.R. Iran
| | - Hadi Bakhtyari
- Department of Biochemistry and Bioinformatics Research Center, School of Pharmacy and Pharmaceutical Sciences, Isfahan University of Medical Sciences, Isfahan, I.R. Iran
| | - Mohammad Reza Mofid
- Department of Biochemistry and Bioinformatics Research Center, School of Pharmacy and Pharmaceutical Sciences, Isfahan University of Medical Sciences, Isfahan, I.R. Iran
| |
Collapse
|
32
|
Abstract
Insulin-like growth factor-binding proteins (IGFBPs) 1-6 bind IGFs but not insulin with high affinity. They were initially identified as serum carriers and passive inhibitors of IGF actions. However, subsequent studies showed that, although IGFBPs inhibit IGF actions in many circumstances, they may also potentiate these actions. IGFBPs are widely expressed in most tissues, and they are flexible endocrine and autocrine/paracrine regulators of IGF activity, which is essential for this important physiological system. More recently, individual IGFBPs have been shown to have IGF-independent actions. Mechanisms underlying these actions include (i) interaction with non-IGF proteins in compartments including the extracellular space and matrix, the cell surface and intracellular space, (ii) interaction with and modulation of other growth factor pathways including EGF, TGF-β and VEGF, and (iii) direct or indirect transcriptional effects following nuclear entry of IGFBPs. Through these IGF-dependent and IGF-independent actions, IGFBPs modulate essential cellular processes including proliferation, survival, migration, senescence, autophagy and angiogenesis. They have been implicated in a range of disorders including malignant, metabolic, neurological and immune diseases. A more complete understanding of their cellular roles may lead to the development of novel IGFBP-based therapeutic opportunities.
Collapse
Affiliation(s)
- L A Bach
- Department of Medicine (Alfred)Monash University, Melbourne, Australia
- Department of Endocrinology and DiabetesAlfred Hospital, Melbourne, Australia
| |
Collapse
|
33
|
Smyth LCD, Rustenhoven J, Park TIH, Schweder P, Jansson D, Heppner PA, O'Carroll SJ, Mee EW, Faull RLM, Curtis M, Dragunow M. Unique and shared inflammatory profiles of human brain endothelia and pericytes. J Neuroinflammation 2018; 15:138. [PMID: 29751771 PMCID: PMC5948925 DOI: 10.1186/s12974-018-1167-8] [Citation(s) in RCA: 74] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2018] [Accepted: 04/18/2018] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Pericytes and endothelial cells are critical cellular components of the blood-brain barrier (BBB) and play an important role in neuroinflammation. To date, the majority of inflammation-related studies in endothelia and pericytes have been carried out using immortalised cell lines or non-human-derived cells. Whether these are representative of primary human cells is unclear and systematic comparisons of the inflammatory responses of primary human brain-derived pericytes and endothelia has yet to be performed. METHODS To study the effects of neuroinflammation at the BBB, primary brain endothelial cells and pericytes were isolated from human biopsy tissue. Culture purity was examined using qPCR and immunocytochemistry. Electrical cell-substrate impedance sensing (ECIS) was used to determine the barrier properties of endothelial and pericyte cultures. Using immunocytochemistry, cytometric bead array, and ECIS, we compared the responses of endothelia and pericytes to a panel of inflammatory stimuli (IL-1β, TNFα, LPS, IFN-γ, TGF-β1, IL-6, and IL-4). Secretome analysis was performed to identify unique secretions of endothelia and pericytes in response to IL-1β. RESULTS Endothelial cells were pure, moderately proliferative, retained the expression of BBB-related junctional proteins and transporters, and generated robust TEER. Both endothelia and pericytes have the same pattern of transcription factor activation in response to inflammatory stimuli but respond differently at the secretion level. Secretome analysis confirmed that endothelia and pericytes have overlapping but distinct secretome profiles in response to IL-1β. We identified several cell-type specific responses, including G-CSF and GM-CSF (endothelial-specific), and IGFBP2 and IGFBP3 (pericyte-specific). Finally, we demonstrated that direct addition of IL-1β, TNFα, LPS, and IL-4 contributed to the loss of endothelial barrier integrity in vitro. CONCLUSIONS Here, we identify important cell-type differences in the inflammatory response of brain pericytes and endothelia and provide, for the first time, a comprehensive profile of the secretions of primary human brain endothelia and pericytes which has implications for understanding how inflammation affects the cerebrovasculature.
Collapse
Affiliation(s)
- Leon C D Smyth
- Department of Pharmacology and Clinical Pharmacology, The University of Auckland, Private Bag 92019, Auckland, 1142, New Zealand.,Centre for Brain Research, The University of Auckland, Private Bag 92019, Auckland, 1142, New Zealand
| | - Justin Rustenhoven
- Department of Pharmacology and Clinical Pharmacology, The University of Auckland, Private Bag 92019, Auckland, 1142, New Zealand.,Centre for Brain Research, The University of Auckland, Private Bag 92019, Auckland, 1142, New Zealand
| | - Thomas I-H Park
- Department of Pharmacology and Clinical Pharmacology, The University of Auckland, Private Bag 92019, Auckland, 1142, New Zealand.,Centre for Brain Research, The University of Auckland, Private Bag 92019, Auckland, 1142, New Zealand.,Department of Anatomy and Medical Imaging, The University of Auckland, Private Bag 92019, Auckland, 1142, New Zealand
| | - Patrick Schweder
- Centre for Brain Research, The University of Auckland, Private Bag 92019, Auckland, 1142, New Zealand.,Auckland City Hospital, Auckland, 1023, New Zealand
| | - Deidre Jansson
- Department of Pharmacology and Clinical Pharmacology, The University of Auckland, Private Bag 92019, Auckland, 1142, New Zealand.,Centre for Brain Research, The University of Auckland, Private Bag 92019, Auckland, 1142, New Zealand
| | - Peter A Heppner
- Centre for Brain Research, The University of Auckland, Private Bag 92019, Auckland, 1142, New Zealand.,Auckland City Hospital, Auckland, 1023, New Zealand
| | - Simon J O'Carroll
- Centre for Brain Research, The University of Auckland, Private Bag 92019, Auckland, 1142, New Zealand.,Department of Anatomy and Medical Imaging, The University of Auckland, Private Bag 92019, Auckland, 1142, New Zealand
| | - Edward W Mee
- Centre for Brain Research, The University of Auckland, Private Bag 92019, Auckland, 1142, New Zealand.,Auckland City Hospital, Auckland, 1023, New Zealand
| | - Richard L M Faull
- Centre for Brain Research, The University of Auckland, Private Bag 92019, Auckland, 1142, New Zealand.,Department of Anatomy and Medical Imaging, The University of Auckland, Private Bag 92019, Auckland, 1142, New Zealand
| | - Maurice Curtis
- Centre for Brain Research, The University of Auckland, Private Bag 92019, Auckland, 1142, New Zealand.,Department of Anatomy and Medical Imaging, The University of Auckland, Private Bag 92019, Auckland, 1142, New Zealand
| | - Mike Dragunow
- Department of Pharmacology and Clinical Pharmacology, The University of Auckland, Private Bag 92019, Auckland, 1142, New Zealand. .,Centre for Brain Research, The University of Auckland, Private Bag 92019, Auckland, 1142, New Zealand.
| |
Collapse
|
34
|
Xiang Q, Liao Y, Chao H, Huang W, Liu J, Chen H, Hong D, Zou Z, Xiang AP, Li W. ISL1 overexpression enhances the survival of transplanted human mesenchymal stem cells in a murine myocardial infarction model. Stem Cell Res Ther 2018; 9:51. [PMID: 29482621 PMCID: PMC5828309 DOI: 10.1186/s13287-018-0803-7] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2017] [Revised: 02/08/2018] [Accepted: 02/08/2018] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND The LIM-homeobox transcription factor islet-1 (ISL1) has been proposed as a marker for cardiovascular progenitor cells. This study investigated whether forced expression of ISL1 in human mesenchymal stem cells (hMSCs) improves myocardial infarction (MI) treatment outcomes. METHODS The lentiviral vector containing the human elongation factor 1α promoter, which drives the expression of ISL1 (EF1α-ISL1), was constructed using the Multisite Gateway System and used to transduce hMSCs. Flow cytometry, immunofluorescence, Western blotting, TUNEL assay, and RNA sequencing were performed to evaluate the function of ISL1-overexpressing hMSCs (ISL1-hMSCs). RESULTS The in vivo results showed that transplantation of ISL1-hMSCs improved cardiac function in a rat model of MI. Left ventricle ejection fraction and fractional shortening were greater in post-MI hearts after 4 weeks of treatment with ISL1-hMSCs compared with control hMSCs or phosphate-buffered saline. We also found that ISL1 overexpression increased angiogenesis and decreased apoptosis and inflammation. The greater potential of ISL1-hMSCs may be attributable to an increased number of surviving cells after transplantation. Conditioned medium from ISL1-hMSCs decreased the apoptotic effect of H2O2 on the cardiomyocyte cell line H9c2. To clarify the molecular basis of this finding, we employed RNA sequencing to compare the apoptotic-related gene expression profiles of control hMSCs and ISL1-hMSCs. The results showed that insulin-like growth factor binding protein 3 (IGFBP3) was the only gene in ISL1-hMSCs with a RPKM value higher than 100 and that the difference fold-change between ISL1-hMSCs and control hMSCs was greater than 3, suggesting that IGFBP3 might play an important role in the anti-apoptosis effect of ISL1-hMSCs through paracrine effects. Furthermore, the expression of IGFBP3 in the conditioned medium from ISL1-hMSCs was almost fourfold greater than that in conditioned medium from control hMSCs. Moreover, the IGFBP3 neutralization antibody reversed the apoptotic effect of ISL1-hMSCs-CM. CONCLUSIONS These results suggest that overexpression of ISL1 in hMSCs promotes cell survival in a model of MI and enhances their paracrine function to protect cardiomyocytes, which may be mediated through IGFBP3. ISL1 overexpression in hMSCs may represent a novel strategy for enhancing the effectiveness of stem cell therapy after MI.
Collapse
Affiliation(s)
- Qiuling Xiang
- Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-sen University, Guangzhou, People's Republic of China.,Zhongshan Medical School, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Yan Liao
- Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-sen University, Guangzhou, People's Republic of China.,Zhongshan Medical School, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Hua Chao
- Zhongshan Medical School, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Weijun Huang
- Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-sen University, Guangzhou, People's Republic of China.,Zhongshan Medical School, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Jia Liu
- Department of Cardiology, the Red Cross hospital of Guangzhou City, the Fourth Affiliated Hospital of Jinan University, Guangzhou, People's Republic of China
| | - Haixuan Chen
- The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Dongxi Hong
- Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Zhengwei Zou
- Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-sen University, Guangzhou, People's Republic of China.,Zhongshan Medical School, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Andy Peng Xiang
- Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-sen University, Guangzhou, People's Republic of China.,Zhongshan Medical School, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Weiqiang Li
- Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-sen University, Guangzhou, People's Republic of China. .,Zhongshan Medical School, Sun Yat-sen University, Guangzhou, People's Republic of China.
| |
Collapse
|
35
|
Abstract
Insulinlike growth factor (IGF) binding proteins (IGFBPs) 1 to 6 are high-affinity regulators of IGF activity. They generally inhibit IGF actions by preventing binding to the IGF-I receptor but can also enhance their actions under some conditions. Posttranslational modifications such as glycosylation and phosphorylation modulate IGFBP properties, and IGFBP proteolysis results in IGF release. IGFBPs have more recently been shown to have IGF-independent actions. A number of mechanisms are involved, including modulation of other growth factor pathways, nuclear localization and transcriptional regulation, interaction with the sphingolipid pathway, and binding to non-IGF biomolecules in the extracellular space and matrix, on the cell surface and intracellularly. IGFBPs modulate important biological processes, including cell proliferation, survival, migration, senescence, autophagy, and angiogenesis. Their actions have been implicated in growth, metabolism, cancer, stem cell maintenance and differentiation, and immune regulation. Recent studies have shown that epigenetic mechanisms are involved in the regulation of IGFBP abundance. A more complete understanding of IGFBP biology is necessary to further define their cellular roles and determine their therapeutic potential.
Collapse
Affiliation(s)
- Leon A Bach
- Department of Endocrinology and Diabetes, The Alfred Hospital, Melbourne, Victoria, Australia
- Department of Medicine, Alfred Medical Research and Education Precinct, Monash University, Melbourne, Victoria, Australia
| |
Collapse
|
36
|
Azam AB, Azizan EAB. Brief Overview of a Decade of Genome-Wide Association Studies on Primary Hypertension. Int J Endocrinol 2018; 2018:7259704. [PMID: 29666641 PMCID: PMC5831899 DOI: 10.1155/2018/7259704] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/28/2017] [Accepted: 12/12/2017] [Indexed: 12/29/2022] Open
Abstract
Primary hypertension is widely believed to be a complex polygenic disorder with the manifestation influenced by the interactions of genomic and environmental factors making identification of susceptibility genes a major challenge. With major advancement in high-throughput genotyping technology, genome-wide association study (GWAS) has become a powerful tool for researchers studying genetically complex diseases. GWASs work through revealing links between DNA sequence variation and a disease or trait with biomedical importance. The human genome is a very long DNA sequence which consists of billions of nucleotides arranged in a unique way. A single base-pair change in the DNA sequence is known as a single nucleotide polymorphism (SNP). With the help of modern genotyping techniques such as chip-based genotyping arrays, thousands of SNPs can be genotyped easily. Large-scale GWASs, in which more than half a million of common SNPs are genotyped and analyzed for disease association in hundreds of thousands of cases and controls, have been broadly successful in identifying SNPs associated with heart diseases, diabetes, autoimmune diseases, and psychiatric disorders. It is however still debatable whether GWAS is the best approach for hypertension. The following is a brief overview on the outcomes of a decade of GWASs on primary hypertension.
Collapse
Affiliation(s)
- Afifah Binti Azam
- Department of Medicine, The National University of Malaysia Medical Centre, Kuala Lumpur, Malaysia
| | - Elena Aisha Binti Azizan
- Department of Medicine, The National University of Malaysia Medical Centre, Kuala Lumpur, Malaysia
| |
Collapse
|
37
|
Stark A, Dammann C, Nielsen HC, Volpe MV. A Pathogenic Relationship of Bronchopulmonary Dysplasia and Retinopathy of Prematurity? A Review of Angiogenic Mediators in Both Diseases. Front Pediatr 2018; 6:125. [PMID: 29951473 PMCID: PMC6008318 DOI: 10.3389/fped.2018.00125] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/06/2017] [Accepted: 04/16/2018] [Indexed: 01/11/2023] Open
Abstract
Bronchopulmonary dysplasia (BPD) and retinopathy of prematurity (ROP) are common and significant morbidities of prematurely born infants. These diseases have in common altered and pathologic vascular formation in the face of incomplete organ development. Therefore, it is reasonable to question whether factors affecting angiogenesis could have a joint pathogenic role for both diseases. Inhibition or induced expression of a single angiogenic factor is unlikely to be 100% causative or protective of either of BPD or ROP. It is more likely that interactions of multiple factors leading to disordered angiogenesis are present, increasing the likelihood of common pathways in both diseases. This review explores this possibility by assessing the evidence showing involvement of specific angiogenic factors in the vascular development and maldevelopment in each disease. Theoretical interactions of specific factors mutually contributing to BPD and ROP are proposed and, where possible, a timeline of the proposed relationships between BPD and ROP is developed. It is hoped that future research will be inspired by the theories put forth in this review to enhance the understanding of the pathogenesis in both diseases.
Collapse
Affiliation(s)
- Ashley Stark
- Tufts University School of Medicine, Boston, MA, United States
| | - Christiane Dammann
- Tufts University School of Medicine, Boston, MA, United States.,Division of Newborn Medicine, Department of Pediatrics, Floating Hospital for Children at Tufts Medical Center, Boston, MA, United States.,Program in Cell, Molecular and Developmental Biology, Sackler School of Graduate Biomedical Sciences, Tufts University School of Medicine, Boston, MA, United States
| | - Heber C Nielsen
- Tufts University School of Medicine, Boston, MA, United States.,Division of Newborn Medicine, Department of Pediatrics, Floating Hospital for Children at Tufts Medical Center, Boston, MA, United States.,Program in Cell, Molecular and Developmental Biology, Sackler School of Graduate Biomedical Sciences, Tufts University School of Medicine, Boston, MA, United States
| | - MaryAnn V Volpe
- Tufts University School of Medicine, Boston, MA, United States.,Division of Newborn Medicine, Department of Pediatrics, Floating Hospital for Children at Tufts Medical Center, Boston, MA, United States
| |
Collapse
|
38
|
Holm M, Morken TS, Fichorova RN, VanderVeen DK, Allred EN, Dammann O, Leviton A. Systemic Inflammation-Associated Proteins and Retinopathy of Prematurity in Infants Born Before the 28th Week of Gestation. Invest Ophthalmol Vis Sci 2017; 58:6419-6428. [PMID: 29260199 PMCID: PMC5736326 DOI: 10.1167/iovs.17-21931] [Citation(s) in RCA: 55] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Purpose To assess the association between systemic levels of inflammation-associated proteins and severe retinopathy of prematurity (ROP) in extremely preterm infants. Methods We collected whole blood on filter paper on postnatal days 1, 7, 14, 21, and 28 from 1205 infants born before the 28th week of gestation, and measured the concentrations of 27 inflammation-associated, angiogenic, and neurotrophic proteins. We calculated odds ratios with 95% confidence intervals for the association between top quartile concentrations of each protein and prethreshold ROP. Results During the first three weeks after birth, high concentrations of VEGF-R1, myeloperoxidase (MPO), IL-8, intercellular adhesion molecule (ICAM)-1, matrix metalloproteinase 9, erythropoietin, TNF-α, and basic fibroblast growth factor were associated with an increased risk for prethreshold ROP. On day 28, high levels of serum amyloid A, MPO, IL-6, TNF-α, TNF-R1/-R2, IL-8, and ICAM-1 were associated with an increased risk. Top quartile concentrations of the proinflammatory cytokines TNF-α and IL-6 were associated with increased risks of ROP when levels of neuroprotective proteins and growth factors, including BDNF, insulin-like growth factor 1, IGFBP-1, VEGFR-1 and -2, ANG-1 and PlGF, were not in the top quartile. In contrast, high concentrations of NT-4 and BDNF appeared protective only in infants without elevated inflammatory mediators. Conclusions Systemic inflammation during the first postnatal month was associated with an increased risk of prethreshold ROP. Elevated concentrations of growth factors, angiogenic proteins, and neurotrophins appeared to modulate this risk, and were capable of reducing the risk even in the absence of systemic inflammation.
Collapse
Affiliation(s)
- Mari Holm
- Department of Clinical and Molecular Medicine, Faculty of Medicine, Norwegian University of Science and Technology, Trondheim, Norway.,Department of Pediatrics, St. Olavs Hospital, Trondheim University Hospital, Trondheim, Norway
| | - Tora S Morken
- Department of Neuromedicine and Movement Science (INB), Faculty of Medicine, Norwegian University of Science and Technology, Trondheim, Norway.,Department of Ophthalmology, St. Olavs Hospital, Trondheim University Hospital, Trondheim, Norway
| | - Raina N Fichorova
- Laboratory of Genital Tract Biology, Department of Obstetrics, Gynecology, and Reproductive Biology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, United States
| | - Deborah K VanderVeen
- Department of Ophthalmology, Children's Hospital Boston, Boston, Massachusetts, United States.,Department of Ophthalmology, Harvard Medical School, Harvard University, Boston, Massachusetts, United States
| | - Elizabeth N Allred
- Department of Neurology, Boston Children's Hospital, Boston, Massachusetts, United States.,Department of Neurology, Harvard Medical School, Harvard University, Boston, Massachusetts, United States
| | - Olaf Dammann
- Department of Public Health and Community Medicine, Tufts University School of Medicine, Boston, Massachusetts, United States.,Perinatal Epidemiology Unit, Department of Gynecology and Obstetrics, Hannover Medical School, Hannover, Germany
| | - Alan Leviton
- Department of Neurology, Boston Children's Hospital, Boston, Massachusetts, United States.,Department of Neurology, Harvard Medical School, Harvard University, Boston, Massachusetts, United States
| | | |
Collapse
|
39
|
Singh JN, Nowlin TM, Seedorf GJ, Abman SH, Shepherd DP. Quantifying three-dimensional rodent retina vascular development using optical tissue clearing and light-sheet microscopy. JOURNAL OF BIOMEDICAL OPTICS 2017; 22:76011. [PMID: 28717817 PMCID: PMC5514054 DOI: 10.1117/1.jbo.22.7.076011] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/21/2017] [Accepted: 06/23/2017] [Indexed: 05/03/2023]
Abstract
Retinal vasculature develops in a highly orchestrated three-dimensional (3-D) sequence. The stages of retinal vascularization are highly susceptible to oxygen perturbations. We demonstrate that optical tissue clearing of intact rat retinas and light-sheet microscopy provides rapid 3-D characterization of vascular complexity during retinal development. Compared with flat mount preparations that dissect the retina and primarily image the outermost vascular layers, intact cleared retinas imaged using light-sheet fluorescence microscopy display changes in the 3-D retinal vasculature rapidly without the need for point scanning techniques. Using a severe model of retinal vascular disruption, we demonstrate that a simple metric based on Sholl analysis captures the vascular changes observed during retinal development in 3-D. Taken together, these results provide a methodology for rapidly quantifying the 3-D development of the entire rodent retinal vasculature.
Collapse
Affiliation(s)
- Jasmine N. Singh
- University of Colorado Denver, Department of Physics, Denver, Colorado, United States
- University of Colorado Anschutz Medical Campus, Pediatric Heart Lung Center, Department of Pediatrics, Aurora, Colorado, United States
| | - Taylor M. Nowlin
- University of Colorado Anschutz Medical Campus, Pediatric Heart Lung Center, Department of Pediatrics, Aurora, Colorado, United States
| | - Gregory J. Seedorf
- University of Colorado Anschutz Medical Campus, Pediatric Heart Lung Center, Department of Pediatrics, Aurora, Colorado, United States
| | - Steven H. Abman
- University of Colorado Anschutz Medical Campus, Pediatric Heart Lung Center, Department of Pediatrics, Aurora, Colorado, United States
| | - Douglas P. Shepherd
- University of Colorado Denver, Department of Physics, Denver, Colorado, United States
- University of Colorado Anschutz Medical Campus, Pediatric Heart Lung Center, Department of Pediatrics, Aurora, Colorado, United States
- Address all correspondence to: Douglas P. Shepherd, E-mail:
| |
Collapse
|
40
|
Baroncelli L, Cenni MC, Melani R, Deidda G, Landi S, Narducci R, Cancedda L, Maffei L, Berardi N. Early IGF-1 primes visual cortex maturation and accelerates developmental switch between NKCC1 and KCC2 chloride transporters in enriched animals. Neuropharmacology 2017; 113:167-177. [PMID: 26924708 DOI: 10.1016/j.neuropharm.2016.02.034] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2015] [Revised: 02/01/2016] [Accepted: 02/24/2016] [Indexed: 12/17/2022]
Abstract
Environmental enrichment (EE) has a remarkable impact on brain development. Continuous exposure to EE from birth determines a significant acceleration of visual system maturation both at retinal and cortical levels. A pre-weaning enriched experience is sufficient to trigger the accelerated maturation of the visual system, suggesting that factors affected by EE during the first days of life might prime visual circuits towards a faster development. The search for such factors is crucial not only to gain a better understanding of the molecular hierarchy of brain development but also to identify molecular pathways amenable to be targeted to correct atypical brain developmental trajectories. Here, we showed that IGF-1 levels are increased in the visual cortex of EE rats as early as P6 and this is a crucial event for setting in motion the developmental program induced by EE. Early intracerebroventricular (i.c.v.) infusion of IGF-1 in standard rats was sufficient to mimic the action of EE on visual acuity development, whereas blocking IGF-1 signaling by i.c.v. injections of the IGF-1 receptor antagonist JB1 prevented the deployment of EE effects. Early IGF-1 decreased the ratio between the expression of NKCC1 and KCC2 cation/chloride transporters, and the reversal potential for GABAAR-driven Cl- currents (ECl) was shifted toward more negative potentials, indicating that IGF-1 is a crucial factor in accelerating the maturation of GABAergic neurotransmission and promoting the developmental switch of GABA polarity from excitation to inhibition. In addition, early IGF-1 promoted a later occurring increase in its own expression, suggesting a priming effect of early IGF-1 in driving post-weaning cortical maturation.
Collapse
Affiliation(s)
- Laura Baroncelli
- Institute of Neuroscience CNR, via Moruzzi 1, I-56124, Pisa, Italy.
| | | | - Riccardo Melani
- Institute of Neuroscience CNR, via Moruzzi 1, I-56124, Pisa, Italy; Department of Neuroscience, Psychology, Drug Research, Child Health (NEUROFARBA), University of Florence, Piazza San Marco 4, I-50121, Florence, Italy
| | - Gabriele Deidda
- Department of Neuroscience and Brain Technologies, Istituto Italiano di Tecnologia, Via Morego 30, I-16163, Genoa, Italy
| | - Silvia Landi
- Institute of Neuroscience CNR, via Moruzzi 1, I-56124, Pisa, Italy
| | - Roberta Narducci
- Department of Neuroscience, Psychology, Drug Research, Child Health (NEUROFARBA), University of Florence, Piazza San Marco 4, I-50121, Florence, Italy
| | - Laura Cancedda
- Department of Neuroscience and Brain Technologies, Istituto Italiano di Tecnologia, Via Morego 30, I-16163, Genoa, Italy
| | - Lamberto Maffei
- Institute of Neuroscience CNR, via Moruzzi 1, I-56124, Pisa, Italy; Laboratory of Neurobiology, Scuola Normale Superiore, Piazza Cavalieri 7, I-56126, Pisa, Italy
| | - Nicoletta Berardi
- Institute of Neuroscience CNR, via Moruzzi 1, I-56124, Pisa, Italy; Department of Neuroscience, Psychology, Drug Research, Child Health (NEUROFARBA), University of Florence, Piazza San Marco 4, I-50121, Florence, Italy
| |
Collapse
|
41
|
Sakimoto S, Marchetti V, Aguilar E, Lee K, Usui Y, Murinello S, Bucher F, Trombley JK, Fallon R, Wagey R, Peters C, Scheppke EL, Westenskow PD, Friedlander M. CD44 expression in endothelial colony-forming cells regulates neurovascular trophic effect. JCI Insight 2017; 2:e89906. [PMID: 28138561 PMCID: PMC5256141 DOI: 10.1172/jci.insight.89906] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Vascular abnormalities are a common component of eye diseases that often lead to vision loss. Vaso-obliteration is associated with inherited retinal degenerations, since photoreceptor atrophy lowers local metabolic demands and vascular support to those regions is no longer required. Given the degree of neurovascular crosstalk in the retina, it may be possible to use one cell type to rescue another cell type in the face of severe stress, such as hypoxia or genetically encoded cell-specific degenerations. Here, we show that intravitreally injected human endothelial colony-forming cells (ECFCs) that can be isolated and differentiated from cord blood in xeno-free media collect in the vitreous cavity and rescue vaso-obliteration and neurodegeneration in animal models of retinal disease. Furthermore, we determined that a subset of the ECFCs was more effective at anatomically and functionally preventing retinopathy; these cells expressed high levels of CD44, the hyaluronic acid receptor, and IGFBPs (insulin-like growth factor-binding proteins). Injection of cultured media from ECFCs or only recombinant human IGFBPs also rescued the ischemia phenotype. These results help us to understand the mechanism of ECFC-based therapies for ischemic insults and retinal neurodegenerative diseases.
Collapse
Affiliation(s)
- Susumu Sakimoto
- Department of Cell and Molecular Biology, The Scripps Research Institute, La Jolla, California, USA
| | | | - Edith Aguilar
- Department of Cell and Molecular Biology, The Scripps Research Institute, La Jolla, California, USA
| | - Kelsey Lee
- STEMCELL Technologies, Vancouver, British Columbia, Canada
| | - Yoshihiko Usui
- Department of Cell and Molecular Biology, The Scripps Research Institute, La Jolla, California, USA
| | - Salome Murinello
- Department of Cell and Molecular Biology, The Scripps Research Institute, La Jolla, California, USA
| | - Felicitas Bucher
- Department of Cell and Molecular Biology, The Scripps Research Institute, La Jolla, California, USA
| | | | - Regis Fallon
- Department of Cell and Molecular Biology, The Scripps Research Institute, La Jolla, California, USA
| | - Ravenska Wagey
- STEMCELL Technologies, Vancouver, British Columbia, Canada
| | - Carrie Peters
- STEMCELL Technologies, Vancouver, British Columbia, Canada
| | | | | | - Martin Friedlander
- Department of Cell and Molecular Biology, The Scripps Research Institute, La Jolla, California, USA
| |
Collapse
|
42
|
Can a Risk Factor Based Approach Safely Reduce Screening for Retinopathy of Prematurity? Int J Pediatr 2017; 2017:9372539. [PMID: 28163726 PMCID: PMC5253483 DOI: 10.1155/2017/9372539] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/19/2016] [Revised: 10/18/2016] [Accepted: 11/13/2016] [Indexed: 11/18/2022] Open
Abstract
Objective. Current American retinopathy of prematurity (ROP) screening guidelines is imprecise for infants ≥ 30 weeks with birth weights between 1500 and 2000 g. Our objective was to evaluate a risk factor based approach for screening premature infants at low risk for severe ROP. Study Design. We performed a 13-year review from Intermountain Health Care (IHC) data. All neonates born at ≤32 weeks were reviewed to determine ROP screening and/or development of severe ROP. Severe ROP was defined by stage ≥ 3 or need for laser therapy. Regression analysis was used to identify significant risk factors for severe ROP. Results. We identified 4607 neonates ≤ 32 weeks gestation. Following exclusion for death, with no retinal exam or incomplete data, 2791 (61%) were included in the study. Overall, severe ROP occurred in 260 (9.3%), but only 11/1601 ≥ 29 weeks (0.7%). All infants with severe ROP ≥ 29 weeks had at least 2 identified ROP risk factors. Implementation of this risk based screening strategy to the IHC population over the timeline of this study would have eliminated screening in 21% (343/1601) of the screened population. Conclusions. Limiting ROP screening for infants ≥ 29 and ≤ 32 weeks to only those with clinical risk factors could significantly reduce screening exams while identifying all infants with severe ROP.
Collapse
|
43
|
Hellström A, Ley D, Hallberg B, Löfqvist C, Hansen-Pupp I, Ramenghi LA, Borg J, Smith LEH, Hård AL. IGF-1 as a Drug for Preterm Infants: A Step-Wise Clinical Development. Curr Pharm Des 2017; 23:5964-5970. [PMID: 28969546 PMCID: PMC5824464 DOI: 10.2174/1381612823666171002114545] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2017] [Accepted: 09/28/2017] [Indexed: 11/22/2022]
Abstract
BACKGROUND Insulin-like growth factor 1 (IGF-1) is a mitogenic hormone involved in many processes such as growth, metabolism, angiogenesis and differentiation. After very preterm birth, energy demands increase while maternal supplies of nutrients and other factors are lost and the infant may become dependent on parenteral nutrition for weeks. Low postnatal IGF-1 concentrations in preterm infants are associated with poor weight gain, retinopathy of prematurity (ROP) and other morbidities. We will describe the process by which we aim to develop supplementation with recombinant human (rh) IGF-1 and its binding protein rhIGFBP-3 as a possible therapy to promote growth and maturation and reduce morbidities in extremely preterm infants. METHODS In order to calculate a dose of IGF-1 tolerated by neonates, a pharmacokinetic study of transfusion with fresh frozen plasma was performed, which provided a relatively low dose of IGF-1, (on average 1.4 µg/kg), that increased serum IGF-1 to levels close to those observed in fetuses and preterm infants of similar GAs. Thereafter, a Phase I 3 hours IV infusion of rhIGF-1/rhIGFBP-3 was conducted in 5 infants, followed by a Phase II study with four sections (A-D). In the Phase II, sections A-D studies, time on infusion increased and younger gestational ages were included. RESULTS IV infusion increased IGF-1 but with short half-life (0.5h) implying a need for continuous infusion. In order to obtain in utero levels of IGF-I, the dose was increased from 100 to 250 µg/kg/24 h and the infusion was prolonged from 3 weeks postnatal age until a postmenstrual age of 29 weeks and 6 days. CONCLUSION The purpose has been to ensure high-quality research into the development of a new drug for preterm infants. We hope that our work will help to establish a new standard for the testing of medications for preterm infants.
Collapse
Affiliation(s)
- Ann Hellström
- Department of Ophthalmology, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - David Ley
- Department of Pediatrics, Institute of Clinical Sciences Lund, Lund University and Skane University Hospital, Lund, Sweden
| | - Boubou Hallberg
- Department of Neonatology, Karolinska Institute and University Hospital, Stockholm, Sweden
| | - Chatarina Löfqvist
- Department of Ophthalmology, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Ingrid Hansen-Pupp
- Department of Pediatrics, Institute of Clinical Sciences Lund, Lund University and Skane University Hospital, Lund, Sweden
| | - Luca A. Ramenghi
- Genova Neonatal Intensive Care Unit, Instituto Pediatrico Giannina Gaslini, GenovaItaly
| | - Jan Borg
- Former Premacure AB, Uppsala, Sweden
| | - Lois E. H. Smith
- Department of Ophthalmology, Boston Children’s Hospital, Harvard Medical School, Boston, MA
| | - Anna-Lena Hård
- Department of Ophthalmology, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| |
Collapse
|
44
|
Cheng GS, Zhang YS, Zhang TT, He L, Wang XY. Bone marrow-derived mesenchymal stem cells modified with IGFBP-3 inhibit the proliferation of pulmonary artery smooth muscle cells. Int J Mol Med 2016; 39:223-230. [PMID: 27959432 DOI: 10.3892/ijmm.2016.2820] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2015] [Accepted: 11/10/2016] [Indexed: 11/06/2022] Open
Abstract
Pulmonary arterial hypertension (PAH) is a common clinical cardiovascular disease, leading to the excessive proliferation of pulmonary artery smooth muscle cells (PASMCs) and endothelial cells, and is associated with a high mortality rate. Recently, stem- and progenitor cell-mediated gene therapies have provided a novel approach for the treatment of PAH. However, the function of human bone marrow-derived mesenchymal stem cells (hBM‑MSCs) modified with the insulin-like growth factor binding protein-3 (IGFBP-3) gene in the regulation of PAH is not yet fully understood. In this study, we explored the biological role of IGFBP‑3-modified hBM‑MSCs in the proliferation of human PASMCs (hPASMCs), and also investigated the potential underlying molecular mechanisms. Our results revealed that IGFBP-3-modified hBM‑MSCs inhibited the proliferation of angiotensin II-stimulated hPASMCs following co-culture on cell culture inserts. In addition, total DNA synthesis and the protein levels of hPASMCs in co-culture were decreased. Moreover, the IGFBP‑3-modified hBM‑MSCs promoted apoptosis and downregulated the expression of B-cell lymphoma-2 (Bcl-2), but increased the expression of Bcl-2 associated X protein (Bax) in hPASMCs. Furthermore, the IGFBP‑3-modified hBM‑MSCs significantly induced a phenotypic change in the hPASMCs from the synthetic to the contractile phenotype in co-culture. Importantly, the levels of several related proteins in the hPASMCs, including phosphorylated (p-)insulin receptor substrate-1 (p-IRS-1), phosphoinositide 3-kinase (p-PI3K), serine/threonine-protein kinase (p-Akt), mitogen-activated protein kinase (p-p38), p-Jun N-terminal kinase (p-JNK) and extracellular signal-regulated kinase (p-ERK), were markedly decreased by the IGFBP-3-modified hBM‑MSCs following co-culture. Taken together, our findings suggest that IGFBP-3-modified hBM‑MSCs inhibit the proliferation and promote the apoptosis of hPASMCs, and promote the swithc to a contractile phenotype in more effectively than wild-type hBM‑MSCs, possibly through the activation of the PI3K/Akt and Ras-mitogen-activated protein kinase (MAPK) signaling pathways. The findings of our study suggest that IGFBP‑3‑modified hBM‑MSCs may be a promising therapeutic strategy for the treatment of PAH.
Collapse
Affiliation(s)
- Ge Sheng Cheng
- Department of Cardiovascular Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Yu Shun Zhang
- Department of Cardiovascular Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Ting Ting Zhang
- Department of Cardiovascular Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Lu He
- Department of Cardiovascular Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Xing Ye Wang
- Department of Cardiovascular Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| |
Collapse
|
45
|
Olivares-González L, Martínez-Fernández de la Cámara C, Hervás D, Marín MP, Lahoz A, Millán JM, Rodrigo R. cGMP-Phosphodiesterase Inhibition Prevents Hypoxia-Induced Cell Death Activation in Porcine Retinal Explants. PLoS One 2016; 11:e0166717. [PMID: 27861632 PMCID: PMC5115799 DOI: 10.1371/journal.pone.0166717] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2016] [Accepted: 11/02/2016] [Indexed: 12/11/2022] Open
Abstract
Retinal hypoxia and oxidative stress are involved in several retinal degenerations including diabetic retinopathy, glaucoma, central retinal artery occlusion, or retinopathy of prematurity. The second messenger cyclic guanosine monophosphate (cGMP) has been reported to be protective for neuronal cells under several pathological conditions including ischemia/hypoxia. The purpose of this study was to evaluate whether the accumulation of cGMP through the pharmacological inhibition of phosphodiesterase (PDE) with Zaprinast prevented retinal degeneration induced by mild hypoxia in cultures of porcine retina. Exposure to mild hypoxia (5% O2) for 24h reduced cGMP content and induced retinal degeneration by caspase dependent and independent (PARP activation) mechanisms. Hypoxia also produced a redox imbalance reducing antioxidant response (superoxide dismutase and catalase activities) and increasing superoxide free radical release. Zaprinast reduced mild hypoxia-induced cell death through inhibition of caspase-3 or PARP activation depending on the cell layer. PDE inhibition also ameliorated the effects of mild hypoxia on antioxidant response and the release of superoxide radical in the photoreceptor layer. The use of a PKG inhibitor, KT5823, suggested that cGMP-PKG pathway is involved in cell survival and antioxidant response. The inhibition of PDE, therefore, could be useful for reducing retinal degeneration under hypoxic/ischemic conditions.
Collapse
Affiliation(s)
- Lorena Olivares-González
- Grupo de Biomedicina Molecular, Celular y Genómica, Instituto de Investigación Sanitaria La Fe, Valencia, Spain
- CIBER de Enfermedades Raras (CIBERER), Madrid, Spain
| | | | - David Hervás
- Unidad de Bioestadística, Instituto de Investigación Sanitaria La Fe, Valencia, Spain
| | - María Pilar Marín
- Unidad de Microscopía, Instituto de Investigación Sanitaria La Fe, Valencia, Spain
| | - Agustin Lahoz
- Unidad de Hepatología Experimental, Unidad Analítica, Instituto de Investigación Sanitaria La Fe, Valencia, Spain
| | - José María Millán
- Grupo de Biomedicina Molecular, Celular y Genómica, Instituto de Investigación Sanitaria La Fe, Valencia, Spain
- CIBER de Enfermedades Raras (CIBERER), Madrid, Spain
| | - Regina Rodrigo
- Grupo de Biomedicina Molecular, Celular y Genómica, Instituto de Investigación Sanitaria La Fe, Valencia, Spain
- CIBER de Enfermedades Raras (CIBERER), Madrid, Spain
- * E-mail:
| |
Collapse
|
46
|
Lee CM, He CH, Nour AM, Zhou Y, Ma B, Park JW, Kim KH, Cruz CD, Sharma L, Nasr ML, Modis Y, Lee CG, Elias JA. IL-13Rα2 uses TMEM219 in chitinase 3-like-1-induced signalling and effector responses. Nat Commun 2016; 7:12752. [PMID: 27629921 PMCID: PMC5027616 DOI: 10.1038/ncomms12752] [Citation(s) in RCA: 77] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2016] [Accepted: 07/28/2016] [Indexed: 12/27/2022] Open
Abstract
Recent studies demonstrated that chitinase 3-like-1 (Chi3l1) binds to and signals via IL-13Rα2. However, the mechanism that IL-13Rα2 uses to mediate the effects of Chi3l1 has not been defined. Here, we demonstrate that the membrane protein, TMEM219, is a binding partner of IL-13Rα2 using yeast two-hybrid, co-immunoprecipitation, co-localization and bimolecular fluorescence complementation assays. Furthermore, fluorescence anisotropy nanodisc assays revealed a direct physical interaction between TMEM219 and IL-13Rα2-Chi3l1 complexes. Null mutations or siRNA silencing of TMEM219 or IL-13Rα2 similarly decreased Chi3l1-stimulated epithelial cell HB-EGF production and macrophage MAPK/Erk and PKB/Akt activation. Null mutations of TMEM219 or IL-13Rα2 also phenocopied one another as regards the ability of Chi3l1 to inhibit oxidant-induced apoptosis and lung injury, promote melanoma metastasis and stimulate TGF-β1. TMEM219 also contributed to the decoy function of IL-13Rα2. These studies demonstrate that TMEM219 plays a critical role in Chi3l1-induced IL-13Rα2 mediated signalling and tissue responses.
Collapse
Affiliation(s)
- Chang-Min Lee
- Department of Molecular Microbiology and Immunology, Brown University, 185 Meeting Street, Box G-L, Providence, Rhode Island 02912, USA
| | - Chuan Hua He
- Department of Molecular Microbiology and Immunology, Brown University, 185 Meeting Street, Box G-L, Providence, Rhode Island 02912, USA
| | - Adel M. Nour
- Department of Molecular Microbiology and Immunology, Brown University, 185 Meeting Street, Box G-L, Providence, Rhode Island 02912, USA
| | - Yang Zhou
- Department of Molecular Microbiology and Immunology, Brown University, 185 Meeting Street, Box G-L, Providence, Rhode Island 02912, USA
| | - Bing Ma
- Department of Molecular Microbiology and Immunology, Brown University, 185 Meeting Street, Box G-L, Providence, Rhode Island 02912, USA
| | - Jin Wook Park
- Department of Molecular Microbiology and Immunology, Brown University, 185 Meeting Street, Box G-L, Providence, Rhode Island 02912, USA
| | - Kyung Hee Kim
- Division of Cardiology, Emory University School of Medicine, Atlanta, Georgia 30322, USA
| | - Charles Dela Cruz
- Section of Pulmonary and Critical Care and Sleep Medicine, Department of Medicine, Yale University School of Medicine, 300 Cedar Street, New Haven, Connecticut 06520, USA
| | - Lokesh Sharma
- Section of Pulmonary and Critical Care and Sleep Medicine, Department of Medicine, Yale University School of Medicine, 300 Cedar Street, New Haven, Connecticut 06520, USA
| | - Mahmoud L. Nasr
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, 240 Longwood Avenue, Boston, Massachusetts 02115, USA
| | - Yorgo Modis
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, 240 Longwood Avenue, Boston, Massachusetts 02115, USA
| | - Chun Geun Lee
- Department of Molecular Microbiology and Immunology, Brown University, 185 Meeting Street, Box G-L, Providence, Rhode Island 02912, USA
| | - Jack A. Elias
- Department of Molecular Microbiology and Immunology, Brown University, 185 Meeting Street, Box G-L, Providence, Rhode Island 02912, USA
- Division of Medicine and Biological Sciences, Warren Alpert School of Medicine, Brown University, Box G-A1, 97 Waterman Street, Providence, Rhode Island 02912, USA
| |
Collapse
|
47
|
Fu Z, Gong Y, Löfqvist C, Hellström A, Smith LEH. Review: adiponectin in retinopathy. BIOCHIMICA ET BIOPHYSICA ACTA 2016; 1862:1392-400. [PMID: 27155572 PMCID: PMC4885769 DOI: 10.1016/j.bbadis.2016.05.002] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/31/2015] [Revised: 03/23/2016] [Accepted: 05/03/2016] [Indexed: 02/06/2023]
Abstract
Neovascular eye diseases are a major cause of blindness including retinopathy of prematurity, diabetic retinopathy and age-related macular degeneration in which new vessel formation is driven by hypoxia or metabolic abnormalities affecting the fuel supply. White-adipose-tissue derived adipokines such as adiponectin modulate metabolic responses. Increasing evidence shows that lack of adiponectin may result in retinal neovascularization. Activation of the adiponectin pathway may in turn restore energy metabolism, to suppress the drive for compensatory but ultimately pathological neovessels of retinopathy. In this review, we will summarize our current knowledge of the role of adiponectin in eye diseases of premature infants, diabetic patients as well as the elderly. Further investigations in this field are likely to lead to new preventative approaches for these diseases.
Collapse
Affiliation(s)
- Zhongjie Fu
- Department of Ophthalmology, Boston Children's Hospital, Harvard Medical School, Boston, MA, United States
| | - Yan Gong
- Department of Ophthalmology, Boston Children's Hospital, Harvard Medical School, Boston, MA, United States
| | - Chatarina Löfqvist
- Department of Ophthalmology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Ann Hellström
- Department of Ophthalmology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Lois E H Smith
- Department of Ophthalmology, Boston Children's Hospital, Harvard Medical School, Boston, MA, United States.
| |
Collapse
|
48
|
Moore GE, Ishida M, Demetriou C, Al-Olabi L, Leon LJ, Thomas AC, Abu-Amero S, Frost JM, Stafford JL, Chaoqun Y, Duncan AJ, Baigel R, Brimioulle M, Iglesias-Platas I, Apostolidou S, Aggarwal R, Whittaker JC, Syngelaki A, Nicolaides KH, Regan L, Monk D, Stanier P. The role and interaction of imprinted genes in human fetal growth. Philos Trans R Soc Lond B Biol Sci 2016; 370:20140074. [PMID: 25602077 PMCID: PMC4305174 DOI: 10.1098/rstb.2014.0074] [Citation(s) in RCA: 101] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Identifying the genetic input for fetal growth will help to understand common, serious complications of pregnancy such as fetal growth restriction. Genomic imprinting is an epigenetic process that silences one parental allele, resulting in monoallelic expression. Imprinted genes are important in mammalian fetal growth and development. Evidence has emerged showing that genes that are paternally expressed promote fetal growth, whereas maternally expressed genes suppress growth. We have assessed whether the expression levels of key imprinted genes correlate with fetal growth parameters during pregnancy, either early in gestation, using chorionic villus samples (CVS), or in term placenta. We have found that the expression of paternally expressing insulin-like growth factor 2 (IGF2), its receptor IGF2R, and the IGF2/IGF1R ratio in CVS tissues significantly correlate with crown–rump length and birthweight, whereas term placenta expression shows no correlation. For the maternally expressing pleckstrin homology-like domain family A, member 2 (PHLDA2), there is no correlation early in pregnancy in CVS but a highly significant negative relationship in term placenta. Analysis of the control of imprinted expression of PHLDA2 gave rise to a maternally and compounded grand-maternally controlled genetic effect with a birthweight increase of 93/155 g, respectively, when one copy of the PHLDA2 promoter variant is inherited. Expression of the growth factor receptor-bound protein 10 (GRB10) in term placenta is significantly negatively correlated with head circumference. Analysis of the paternally expressing delta-like 1 homologue (DLK1) shows that the paternal transmission of type 1 diabetes protective G allele of rs941576 single nucleotide polymorphism (SNP) results in significantly reduced birth weight (−132 g). In conclusion, we have found that the expression of key imprinted genes show a strong correlation with fetal growth and that for both genetic and genomics data analyses, it is important not to overlook parent-of-origin effects.
Collapse
Affiliation(s)
- Gudrun E Moore
- Genetics and Epigenetics in Health and Diseases Section, Genetics and Genomic Medicine Programme, UCL Institute of Child Health, London WC1N 1EH, UK
| | - Miho Ishida
- Genetics and Epigenetics in Health and Diseases Section, Genetics and Genomic Medicine Programme, UCL Institute of Child Health, London WC1N 1EH, UK
| | - Charalambos Demetriou
- Genetics and Epigenetics in Health and Diseases Section, Genetics and Genomic Medicine Programme, UCL Institute of Child Health, London WC1N 1EH, UK
| | - Lara Al-Olabi
- Genetics and Epigenetics in Health and Diseases Section, Genetics and Genomic Medicine Programme, UCL Institute of Child Health, London WC1N 1EH, UK
| | - Lydia J Leon
- Genetics and Epigenetics in Health and Diseases Section, Genetics and Genomic Medicine Programme, UCL Institute of Child Health, London WC1N 1EH, UK
| | - Anna C Thomas
- Genetics and Epigenetics in Health and Diseases Section, Genetics and Genomic Medicine Programme, UCL Institute of Child Health, London WC1N 1EH, UK
| | - Sayeda Abu-Amero
- Genetics and Epigenetics in Health and Diseases Section, Genetics and Genomic Medicine Programme, UCL Institute of Child Health, London WC1N 1EH, UK
| | - Jennifer M Frost
- Genetics and Epigenetics in Health and Diseases Section, Genetics and Genomic Medicine Programme, UCL Institute of Child Health, London WC1N 1EH, UK
| | - Jaime L Stafford
- Genetics and Epigenetics in Health and Diseases Section, Genetics and Genomic Medicine Programme, UCL Institute of Child Health, London WC1N 1EH, UK
| | - Yao Chaoqun
- Genetics and Epigenetics in Health and Diseases Section, Genetics and Genomic Medicine Programme, UCL Institute of Child Health, London WC1N 1EH, UK
| | - Andrew J Duncan
- Genetics and Epigenetics in Health and Diseases Section, Genetics and Genomic Medicine Programme, UCL Institute of Child Health, London WC1N 1EH, UK
| | - Rachel Baigel
- Genetics and Epigenetics in Health and Diseases Section, Genetics and Genomic Medicine Programme, UCL Institute of Child Health, London WC1N 1EH, UK
| | - Marina Brimioulle
- Genetics and Epigenetics in Health and Diseases Section, Genetics and Genomic Medicine Programme, UCL Institute of Child Health, London WC1N 1EH, UK
| | - Isabel Iglesias-Platas
- Genetics and Epigenetics in Health and Diseases Section, Genetics and Genomic Medicine Programme, UCL Institute of Child Health, London WC1N 1EH, UK
| | - Sophia Apostolidou
- Genetics and Epigenetics in Health and Diseases Section, Genetics and Genomic Medicine Programme, UCL Institute of Child Health, London WC1N 1EH, UK
| | - Reena Aggarwal
- Genetics and Epigenetics in Health and Diseases Section, Genetics and Genomic Medicine Programme, UCL Institute of Child Health, London WC1N 1EH, UK
| | - John C Whittaker
- Noncommunicable Disease Epidemiology Unit, London School of Hygiene and Tropical Medicine, University of London, London WC1E 7HT, UK
| | - Argyro Syngelaki
- Harris Birthright Research Centre for Fetal Medicine, King's College Hospital, London SE5 9RS, UK
| | - Kypros H Nicolaides
- Harris Birthright Research Centre for Fetal Medicine, King's College Hospital, London SE5 9RS, UK
| | - Lesley Regan
- Department of Obstetrics and Gynaecology, Imperial College London, St Mary's Campus, London W2 1NY, UK
| | - David Monk
- Genetics and Epigenetics in Health and Diseases Section, Genetics and Genomic Medicine Programme, UCL Institute of Child Health, London WC1N 1EH, UK
| | - Philip Stanier
- Genetics and Epigenetics in Health and Diseases Section, Genetics and Genomic Medicine Programme, UCL Institute of Child Health, London WC1N 1EH, UK
| |
Collapse
|
49
|
Cayabyab R, Ramanathan R. Retinopathy of Prematurity: Therapeutic Strategies Based on Pathophysiology. Neonatology 2016; 109:369-76. [PMID: 27251645 DOI: 10.1159/000444901] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Retinopathy of prematurity (ROP) continues to be a major preventable cause of blindness and visual handicaps globally. With improved perinatal care, improved survival of moderately preterm infants, and limited resources for oxygen delivery and monitoring, more mature preterm infants are developing severe ROP in developing countries. The pathophysiology of ROP is characterized by two phases. Phase I ROP is due to vaso-obliteration beginning immediately after birth secondary to a marked decrease in vascular endothelial growth factor (VEGF) and insulin-like growth factor-1 (IGF-1). Phase II begins around 33 weeks' postmenstrual age (PMA). During this phase, VEGF levels increase, especially if there is retinal hypoxia with increasing retinal metabolism and demand for oxygen leading to abnormal vasoproliferation. Since the original description of ROP in 1942 by Terry et al. [Am J Ophthalmol 1942;25:203-204], four epidemics of ROP have been observed. Prevention or early treatment of ROP involves careful titration of oxygen saturation by pulse oximeter (SpO2). Optimal SpO2 target remains elusive. Most of the large trials have focused on either a low SpO2 (85-89%) or a high SpO2 (91-95%) from the first day of birth to 36 weeks' PMA. Although the incidence of severe ROP and bronchopulmonary dysplasia decreased significantly, predischarge mortality was higher in these studies. Use of graded SpO2 during the 2 different phases of ROP (early, low SpO2 during phase I vs. late, high SpO2 during phase II) may be the best approach to prevent this disabling condition. Further trials should focus on this strategy. Other biological agents that are currently being studied include IGF-1 with IGF-binding protein-3 (rhIGF-1 + rhIGFBP-3) and propranolol. For advanced stages of ROP, laser ablation of avascular retina, early treatment of ROP (ETROP) protocol, intravitreal injection of anti-VEGF antibodies (e.g. bevacizumab) and vitrectomy are used to protect central vision and prevent retinal detachment. Long-term complications such as refractory errors, recurrence of ROP and risk of retinal detachment require continued follow-up with an ophthalmologist through adolescence and beyond. Optimal nutrition including adequate intake of omega-3 polyunsaturated fatty acids and decreasing infection/inflammation to promote normal vascularization are important strategies. Screening guidelines for ROP based on local incidence of ROP in different regions of the world are very important. Oxygen therapy is clearly a modifiable risk factor to decrease ROP that needs further study. Understanding the two phases of ROP will help to identify appropriate therapeutic strategies and improve visual outcomes in many preterm infants globally.
Collapse
Affiliation(s)
- Rowena Cayabyab
- Division of Neonatal Medicine, Department of Pediatrics, LAC+USC Medical Center and Children's Hospital Los Angeles, Keck School of Medicine, University of Southern California, Los Angeles, Calif., USA
| | | |
Collapse
|
50
|
Lee YM, Kim J, Kim CS, Jo K, Yoo NH, Sohn E, Kim JS. Anti-glycation and anti-angiogenic activities of 5'-methoxybiphenyl-3,4,3'-triol, a novel phytochemical component of Osteomeles schwerinae. Eur J Pharmacol 2015; 760:172-8. [PMID: 25917323 DOI: 10.1016/j.ejphar.2015.04.022] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2014] [Revised: 03/09/2015] [Accepted: 04/15/2015] [Indexed: 11/29/2022]
Abstract
Advanced glycation end products (AGEs) are involved in the development of diabetic complications such as diabetic retinopathy. 5'-methoxybiphenyl-3,4,3'-triol (referred to as K24) was isolated using bioactivity-guided fractionation of Osteomeles schwerinae C. K. Schneid. and identified as a potent AGE inhibitor. To identify the protective effect of K24 on disruption of the blood-retinal barrier, AGE-RSA was intravitreally injected into rat eyes. K24 had an inhibitory effect on AGE-RSA-induced retinal vascular leakage by suppressing the expression of vascular endothelial growth factor (VEGF) and decreasing the loss of occludin. In addition, we examined whether K24 has a preventive effect against retinal pathogenic angiogenesis in an oxygen-induced retinopathy (OIR) mouse model. K24 significantly reduced the retinal non-perfused area and neovascular tufts in the OIR mice. These data indicate that K24 could serve as an innovative pharmaceutical agent to prevent blood-retinal barrier breakage and retinal pathogenic angiogenesis through an anti-VEGF mechanism.
Collapse
Affiliation(s)
- Yun Mi Lee
- Korean Medicine Based Herbal Drug Development Group, Herbal Medicine Research Division, Korea Institute of Oriental Medicine, Daejeon, South Korea
| | - Junghyun Kim
- Korean Medicine Based Herbal Drug Development Group, Herbal Medicine Research Division, Korea Institute of Oriental Medicine, Daejeon, South Korea
| | - Chan-Sik Kim
- Korean Medicine Based Herbal Drug Development Group, Herbal Medicine Research Division, Korea Institute of Oriental Medicine, Daejeon, South Korea
| | - Kyuhyung Jo
- Korean Medicine Based Herbal Drug Development Group, Herbal Medicine Research Division, Korea Institute of Oriental Medicine, Daejeon, South Korea
| | - Nam Hee Yoo
- International Ginseng & Herb Research Institute, Geumsan, South Korea
| | - Eunjin Sohn
- Korean Medicine Based Herbal Drug Development Group, Herbal Medicine Research Division, Korea Institute of Oriental Medicine, Daejeon, South Korea
| | - Jin Sook Kim
- Korean Medicine Based Herbal Drug Development Group, Herbal Medicine Research Division, Korea Institute of Oriental Medicine, Daejeon, South Korea.
| |
Collapse
|