1
|
Shahidi M. Quantifying Retinal Oxygenation and Metabolism by Phosphorescence Lifetime Imaging. Exp Eye Res 2025:110422. [PMID: 40381978 DOI: 10.1016/j.exer.2025.110422] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2025] [Revised: 04/15/2025] [Accepted: 05/12/2025] [Indexed: 05/20/2025]
Abstract
The retina is a highly metabolically active tissue, requiring adequate availability of oxygen and other metabolites to generate energy for cellular survival and visual function. Retinal hypoxia has been implicated in several common retinal diseases and associated with the development of vision-threatening pathologies. Since the level of hypoxia determines processes that are activated for either cell survival or death, knowledge of retinal oxygenation is essential. This article reviews depth-resolved quantitative measurements of retinal vascular and tissue oxygen tension in rodents using the technique of phosphorescence lifetime imaging. Furthermore, retinal oxygen metabolic biomarkers were quantitatively derived from oxygen tension measurements and shown to be altered under challenged physiological and pathological conditions. Application of phosphorescence lifetime imaging can be useful for advancing knowledge of retinal ischemia pathophysiology and identifying physiological biomarkers to monitor progression and evaluate therapeutic interventions in animal models of human retinal diseases.
Collapse
Affiliation(s)
- Mahnaz Shahidi
- Department of Ophthalmology, University of Southern California Keck School of Medicine, Los Angeles, CA, United States; Alfred E. Mann Department of Biomedical Engineering, University of Southern California, Los Angeles, CA, USA.
| |
Collapse
|
2
|
Heo JI, Ryu J. Natural Products in the Treatment of Retinopathy of Prematurity: Exploring Therapeutic Potentials. Int J Mol Sci 2024; 25:8461. [PMID: 39126030 PMCID: PMC11313229 DOI: 10.3390/ijms25158461] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2024] [Revised: 07/30/2024] [Accepted: 07/31/2024] [Indexed: 08/12/2024] Open
Abstract
Retinopathy of prematurity (ROP) is a vascular disorder affecting the retinas of preterm infants. This condition arises when preterm infants in incubators are exposed to high oxygen levels, leading to oxidative stress, inflammatory responses, and a downregulation of vascular endothelial growth factors, which causes the loss of retinal microvascular capillaries. Upon returning to room air, the upregulation of vascular growth factors results in abnormal vascular growth of retinal endothelial cells. Without appropriate intervention, ROP can progress to blindness. The prevalence of ROP has risen, making it a significant cause of childhood blindness. Current treatments, such as laser therapy and various pharmacologic approaches, are limited by their potential for severe adverse effects. Therefore, a deeper understanding of ROP's pathophysiology and the development of innovative treatments are imperative. Natural products from plants, fungi, bacteria, and marine organisms have shown promise in treating various diseases and have gained attention in ROP research due to their minimal side effects and wide-ranging beneficial properties. This review discusses the roles and mechanisms of natural products that hold potential as therapeutic agents in ROP management.
Collapse
Affiliation(s)
| | - Juhee Ryu
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Kyungpook National University, Daegu 41566, Republic of Korea;
| |
Collapse
|
3
|
Zhang XT, Zhao BW, Zhang YL, Chen S. Human umbilical cord mesenchymal stem cells derived-exosomes on VEGF-A in hypoxic-induced mice retinal astrocytes and mice model of retinopathy of prematurity. Int J Ophthalmol 2024; 17:1238-1247. [PMID: 39026907 PMCID: PMC11246947 DOI: 10.18240/ijo.2024.07.07] [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: 12/17/2023] [Accepted: 04/01/2024] [Indexed: 07/20/2024] Open
Abstract
AIM To observe the effect of human umbilical cord mesenchymal stem cells (hUCMSCs) secretions on the relevant factors in mouse retinal astrocytes, and to investigate the effect of hUCMSCs on the expression of vascular endothelial growth factor-A (VEGF-A) and to observe the therapeutic effect on the mouse model of retinopathy of prematurity (ROP). METHODS Cultured hUCMSCs and extracted exosomes from them and then retinal astrocytes were divided into control group and hypoxia group. MTT assay, flow cytometry, reverse transcription-polymerase chain reaction (RT-PCR) and Western blot were used to detect related indicators. Possible mechanisms by which hUCMSCs exosomes affect VEGF-A expression in hypoxia-induced mouse retinal astrocytes were explored. At last, the efficacy of exosomes of UCMSCs in a mouse ROP model was explored. Graphpad6 was used to comprehensively process data information. RESULTS The secretion was successfully extracted from the culture supernatant of hUCMSCs by gradient ultracentrifugation. Reactive oxygen species (ROS) and hypoxia inducible factor-1α (HIF-1α) of mice retinal astrocytes under different hypoxia time and the expression level of VEGF-A protein and VEGF-A mRNA increased, and the ROP cell model was established after 6h of hypoxia. The secretions of medium and high concentrations of hUCMSCs can reduce ROS and HIF-1α, the expression levels of VEGF-A protein and VEGF-A mRNA are statistically significant and concentration dependent. Compared with the ROP cell model group, the expression of phosphatidylinositol 3-kinase (PI3K)/protein kinase B (AKT)/mammalian target of rapamycin (mTOR) signal pathway related factors in the hUCMSCs exocrine group is significantly decreased. The intravitreal injection of the secretions of medium and high concentrations of hUCMSCs can reduce VEGF-A and HIF-1α in ROP model tissues. HE staining shows that the number of retinal neovascularization in ROP mice decreases with the increase of the dose of hUCMSCs secretion. CONCLUSION In a hypoxia induced mouse retinal astrocyte model, hUCMSCs exosomes are found to effectively reduce the expression of HIF-1α and VEGF-A, which are positively correlated with the concentration of hUCMSCs exosomes. HUCMSCs exosomes can effectively reduce the number of retinal neovascularization and the expression of HIF-1α and VEGF-A proteins in ROP mice, and are positively correlated with drug dosage. Besides, they can reduce the related factors on the PI3K/AKT/mTOR signaling pathway.
Collapse
Affiliation(s)
- Xiao-Tian Zhang
- Tianjin Eye Hospital, Tianjin 300020, China
- Tianjin Key Lab of Ophthalmology and Visual Science, Tianjin 300020, China
| | - Bo-Wen Zhao
- Tianjin Eye Hospital, Tianjin 300020, China
- Tianjin Key Lab of Ophthalmology and Visual Science, Tianjin 300020, China
| | - Yuan-Long Zhang
- Tianjin Eye Hospital, Tianjin 300020, China
- Tianjin Key Lab of Ophthalmology and Visual Science, Tianjin 300020, China
| | - Song Chen
- Tianjin Eye Hospital, Tianjin 300020, China
- Tianjin Key Lab of Ophthalmology and Visual Science, Tianjin 300020, China
| |
Collapse
|
4
|
Zhang L, Buonfiglio F, Fieß A, Pfeiffer N, Gericke A. Retinopathy of Prematurity-Targeting Hypoxic and Redox Signaling Pathways. Antioxidants (Basel) 2024; 13:148. [PMID: 38397746 PMCID: PMC10885953 DOI: 10.3390/antiox13020148] [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: 12/30/2023] [Revised: 01/19/2024] [Accepted: 01/21/2024] [Indexed: 02/25/2024] Open
Abstract
Retinopathy of prematurity (ROP) is a proliferative vascular ailment affecting the retina. It is the main risk factor for visual impairment and blindness in infants and young children worldwide. If left undiagnosed and untreated, it can progress to retinal detachment and severe visual impairment. Geographical variations in ROP epidemiology have emerged over recent decades, attributable to differing levels of care provided to preterm infants across countries and regions. Our understanding of the causes of ROP, screening, diagnosis, treatment, and associated risk factors continues to advance. This review article aims to present the pathophysiological mechanisms of ROP, including its treatment. Specifically, it delves into the latest cutting-edge treatment approaches targeting hypoxia and redox signaling pathways for this condition.
Collapse
Affiliation(s)
| | | | | | | | - Adrian Gericke
- Department of Ophthalmology, University Medical Center, Johannes Gutenberg University Mainz, Langenbeckstrasse 1, 55131 Mainz, Germany; (L.Z.); (F.B.); (A.F.); (N.P.)
| |
Collapse
|
5
|
Ren R, Ding S, Ma K, Jiang Y, Wang Y, Chen J, Wang Y, Kou Y, Fan X, Zhu X, Qin L, Qiu C, Simons M, Wei X, Yu L. SUMOylation Fine-Tunes Endothelial HEY1 in the Regulation of Angiogenesis. Circ Res 2024; 134:203-222. [PMID: 38166414 PMCID: PMC10872267 DOI: 10.1161/circresaha.123.323398] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Accepted: 12/15/2023] [Indexed: 01/04/2024]
Abstract
BACKGROUND Angiogenesis, which plays a critical role in embryonic development and tissue repair, is controlled by a set of angiogenic signaling pathways. As a TF (transcription factor) belonging to the basic helix-loop-helix family, HEY (hairy/enhancer of split related with YRPW motif)-1 (YRPW motif, abbreviation of 4 highly conserved amino acids in the motif) has been identified as a key player in developmental angiogenesis. However, the precise mechanisms underlying HEY1's actions in angiogenesis remain largely unknown. Our previous studies have suggested a potential role for posttranslational SUMOylation in the dynamic regulation of vascular development and organization. METHODS Immunoprecipitation, mass spectrometry, and bioinformatics analysis were used to determine the biochemical characteristics of HEY1 SUMOylation. The promoter-binding capability of HEY1 was determined by chromatin immunoprecipitation, dual luciferase, and electrophoretic mobility shift assays. The dimerization pattern of HEY1 was determined by coimmunoprecipitation. The angiogenic capabilities of endothelial cells were assessed by CCK-8 (cell counting kit-8), 5-ethynyl-2-deoxyuridine staining, wound healing, transwell, and sprouting assays. Embryonic and postnatal vascular growth in mouse tissues, matrigel plug assay, cutaneous wound healing model, oxygen-induced retinopathy model, and tumor angiogenesis model were used to investigate the angiogenesis in vivo. RESULTS We identified intrinsic endothelial HEY1 SUMOylation at conserved lysines by TRIM28 (tripartite motif containing 28) as the unique E3 ligase. Functionally, SUMOylation facilitated HEY1-mediated suppression of angiogenic RTK (receptor tyrosine kinase) signaling and angiogenesis in primary human endothelial cells and mice with endothelial cell-specific expression of wild-type HEY1 or a SUMOylation-deficient HEY1 mutant. Mechanistically, SUMOylation facilitates HEY1 homodimer formation, which in turn preserves HEY1's DNA-binding capability via recognition of E-box promoter elements. Therefore, SUMOylation maintains HEY1's function as a repressive TF controlling numerous angiogenic genes, including RTKs and Notch pathway components. Proangiogenic stimuli induce HEY1 deSUMOylation, leading to heterodimerization of HEY1 with HES (hairy and enhancer of split)-1, which results in ineffective DNA binding and loss of HEY1's angiogenesis-suppressive activity. CONCLUSIONS Our findings demonstrate that reversible HEY1 SUMOylation is a molecular mechanism that coordinates endothelial angiogenic signaling and angiogenesis, both in physiological and pathological milieus, by fine-tuning the transcriptional activity of HEY1. Specifically, SUMOylation facilitates the formation of the HEY1 transcriptional complex and enhances its DNA-binding capability in endothelial cells.
Collapse
Affiliation(s)
- Ruizhe Ren
- Key Laboratory of Cardiovascular Intervention and Regenerative Medicine of Zhejiang Province of Sir Run Run Shaw Hospital, MOE Laboratory of Biosystems Homeostasis & Protection of College of Life Sciences, Zhejiang University, Hangzhou, Zhejiang, China
- Cancer Center and Joint Research Centre for Engineering Biology, Zhejiang University, Zhejiang, China
| | - Sha Ding
- Key Laboratory of Cardiovascular Intervention and Regenerative Medicine of Zhejiang Province of Sir Run Run Shaw Hospital, MOE Laboratory of Biosystems Homeostasis & Protection of College of Life Sciences, Zhejiang University, Hangzhou, Zhejiang, China
- Cancer Center and Joint Research Centre for Engineering Biology, Zhejiang University, Zhejiang, China
| | - Kefan Ma
- Key Laboratory of Cardiovascular Intervention and Regenerative Medicine of Zhejiang Province of Sir Run Run Shaw Hospital, MOE Laboratory of Biosystems Homeostasis & Protection of College of Life Sciences, Zhejiang University, Hangzhou, Zhejiang, China
- Cancer Center and Joint Research Centre for Engineering Biology, Zhejiang University, Zhejiang, China
| | - Yuanqing Jiang
- Key Laboratory of Cardiovascular Intervention and Regenerative Medicine of Zhejiang Province of Sir Run Run Shaw Hospital, MOE Laboratory of Biosystems Homeostasis & Protection of College of Life Sciences, Zhejiang University, Hangzhou, Zhejiang, China
- Cancer Center and Joint Research Centre for Engineering Biology, Zhejiang University, Zhejiang, China
| | - Yiran Wang
- Key Laboratory of Cardiovascular Intervention and Regenerative Medicine of Zhejiang Province of Sir Run Run Shaw Hospital, MOE Laboratory of Biosystems Homeostasis & Protection of College of Life Sciences, Zhejiang University, Hangzhou, Zhejiang, China
- Cancer Center and Joint Research Centre for Engineering Biology, Zhejiang University, Zhejiang, China
| | - Junbo Chen
- Key Laboratory of Cardiovascular Intervention and Regenerative Medicine of Zhejiang Province of Sir Run Run Shaw Hospital, MOE Laboratory of Biosystems Homeostasis & Protection of College of Life Sciences, Zhejiang University, Hangzhou, Zhejiang, China
- Cancer Center and Joint Research Centre for Engineering Biology, Zhejiang University, Zhejiang, China
| | - Yunyun Wang
- Key Laboratory of Cardiovascular Intervention and Regenerative Medicine of Zhejiang Province of Sir Run Run Shaw Hospital, MOE Laboratory of Biosystems Homeostasis & Protection of College of Life Sciences, Zhejiang University, Hangzhou, Zhejiang, China
- Cancer Center and Joint Research Centre for Engineering Biology, Zhejiang University, Zhejiang, China
| | - Yaohui Kou
- Key Laboratory of Cardiovascular Intervention and Regenerative Medicine of Zhejiang Province of Sir Run Run Shaw Hospital, MOE Laboratory of Biosystems Homeostasis & Protection of College of Life Sciences, Zhejiang University, Hangzhou, Zhejiang, China
- Cancer Center and Joint Research Centre for Engineering Biology, Zhejiang University, Zhejiang, China
| | - Xiao Fan
- Key Laboratory of Cardiovascular Intervention and Regenerative Medicine of Zhejiang Province of Sir Run Run Shaw Hospital, MOE Laboratory of Biosystems Homeostasis & Protection of College of Life Sciences, Zhejiang University, Hangzhou, Zhejiang, China
| | - Xiaolong Zhu
- Key Laboratory of Cardiovascular Intervention and Regenerative Medicine of Zhejiang Province of Sir Run Run Shaw Hospital, MOE Laboratory of Biosystems Homeostasis & Protection of College of Life Sciences, Zhejiang University, Hangzhou, Zhejiang, China
- Cancer Center and Joint Research Centre for Engineering Biology, Zhejiang University, Zhejiang, China
| | - Lingfeng Qin
- Department of Surgery, Program in Vascular Biology and Therapeutics, Yale School of Medicine, New Haven, Connecticut, USA
| | - Cong Qiu
- Key Laboratory of Cardiovascular Intervention and Regenerative Medicine of Zhejiang Province of Sir Run Run Shaw Hospital, MOE Laboratory of Biosystems Homeostasis & Protection of College of Life Sciences, Zhejiang University, Hangzhou, Zhejiang, China
- Cancer Center and Joint Research Centre for Engineering Biology, Zhejiang University, Zhejiang, China
| | - Michael Simons
- Yale Cardiovascular Research Center, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, USA
| | - Xiyang Wei
- Key Laboratory of Cardiovascular Intervention and Regenerative Medicine of Zhejiang Province of Sir Run Run Shaw Hospital, MOE Laboratory of Biosystems Homeostasis & Protection of College of Life Sciences, Zhejiang University, Hangzhou, Zhejiang, China
| | - Luyang Yu
- Key Laboratory of Cardiovascular Intervention and Regenerative Medicine of Zhejiang Province of Sir Run Run Shaw Hospital, MOE Laboratory of Biosystems Homeostasis & Protection of College of Life Sciences, Zhejiang University, Hangzhou, Zhejiang, China
- Cancer Center and Joint Research Centre for Engineering Biology, Zhejiang University, Zhejiang, China
| |
Collapse
|
6
|
Dammann O, Stansfield BK. Neonatal sepsis as a cause of retinopathy of prematurity: An etiological explanation. Prog Retin Eye Res 2024; 98:101230. [PMID: 37984792 PMCID: PMC10842718 DOI: 10.1016/j.preteyeres.2023.101230] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Revised: 11/12/2023] [Accepted: 11/13/2023] [Indexed: 11/22/2023]
Abstract
Retinopathy of prematurity (ROP) is a complex neonatal disorder with multiple contributing factors. In this paper we have mounted the evidence in support of the proposal that neonatal sepsis meets all requirements for being a cause of ROP (not a condition, mechanism, or even innocent bystander) by means of initiating the early stages of the pathomechanism of ROP occurrence, systemic inflammation. We use the model of etiological explanation, which distinguishes between two overlapping processes in ROP causation. It can be shown that sepsis can initiate the early stages of the pathomechanism via systemic inflammation (causation process) and that systemic inflammation can contribute to growth factor aberrations and the retinal characteristics of ROP (disease process). The combined contribution of these factors with immaturity at birth (as intrinsic risk modifier) and prenatal inflammation (as extrinsic facilitator) seems to provide a cogent functional framework of ROP occurrence. Finally, we apply the Bradford Hill heuristics to the available evidence. Taken together, the above suggests that neonatal sepsis is a causal inducer of ROP.
Collapse
Affiliation(s)
- Olaf Dammann
- Dept. of Public Health & Community Medicine, Tufts University School of Medicine, Boston, USA; Dept. of Gynecology & Obstetrics, Hannover Medical School, Hannover, Germany; Dept. of Neuromedicine & Movement Science, Norwegian University of Science & Technology, Trondheim, Norway; Dept. of Philosophy, University of Johannesburg, Johannesburg, South Africa.
| | | |
Collapse
|
7
|
Pérez-Gutiérrez L, Ferrara N. Biology and therapeutic targeting of vascular endothelial growth factor A. Nat Rev Mol Cell Biol 2023; 24:816-834. [PMID: 37491579 DOI: 10.1038/s41580-023-00631-w] [Citation(s) in RCA: 149] [Impact Index Per Article: 74.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/16/2023] [Indexed: 07/27/2023]
Abstract
The formation of new blood vessels, called angiogenesis, is an essential pathophysiological process in which several families of regulators have been implicated. Among these, vascular endothelial growth factor A (VEGFA; also known as VEGF) and its two tyrosine kinase receptors, VEGFR1 and VEGFR2, represent a key signalling pathway mediating physiological angiogenesis and are also major therapeutic targets. VEGFA is a member of the gene family that includes VEGFB, VEGFC, VEGFD and placental growth factor (PLGF). Three decades after its initial isolation and cloning, VEGFA is arguably the most extensively investigated signalling system in angiogenesis. Although many mediators of angiogenesis have been identified, including members of the FGF family, angiopoietins, TGFβ and sphingosine 1-phosphate, all current FDA-approved anti-angiogenic drugs target the VEGF pathway. Anti-VEGF agents are widely used in oncology and, in combination with chemotherapy or immunotherapy, are now the standard of care in multiple malignancies. Anti-VEGF drugs have also revolutionized the treatment of neovascular eye disorders such as age-related macular degeneration and ischaemic retinal disorders. In this Review, we emphasize the molecular, structural and cellular basis of VEGFA action as well as recent findings illustrating unexpected interactions with other pathways and provocative reports on the role of VEGFA in regenerative medicine. We also discuss clinical and translational aspects of VEGFA. Given the crucial role that VEGFA plays in regulating angiogenesis in health and disease, this molecule is largely the focus of this Review.
Collapse
Affiliation(s)
- Lorena Pérez-Gutiérrez
- Department of Pathology, University of California San Diego, La Jolla, CA, USA
- Department of Ophthalmology, University of California San Diego, La Jolla, CA, USA
- Moores Cancer Center, University of California San Diego, La Jolla, CA, USA
| | - Napoleone Ferrara
- Department of Pathology, University of California San Diego, La Jolla, CA, USA.
- Department of Ophthalmology, University of California San Diego, La Jolla, CA, USA.
- Moores Cancer Center, University of California San Diego, La Jolla, CA, USA.
| |
Collapse
|
8
|
Abstract
Retinopathy of prematurity (ROP) is a complex disease involving development of the neural retina, ocular circulations, and other organ systems of the premature infant. The external stresses of the ex utero environment also influence the pathophysiology of ROP through interactions among retinal neural, vascular, and glial cells. There is variability among individual infants and presentations of the disease throughout the world, making ROP challenging to study. The methods used include representative animal models, cell culture, and clinical studies. This article describes the impact of maternal-fetal interactions; stresses that the preterm infant experiences; and biologic pathways of interest, including growth factor effects and cell-cell interactions, on the complex pathophysiology of ROP phenotypes in developed and emerging countries.
Collapse
|
9
|
Zhao Y, Zhong Y, Chen W, Chang S, Cao Q, Wang Y, Yang L. Ocular and neural genes jointly regulate the visuospatial working memory in ADHD children. BEHAVIORAL AND BRAIN FUNCTIONS : BBF 2023; 19:14. [PMID: 37658396 PMCID: PMC10472596 DOI: 10.1186/s12993-023-00216-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/23/2023] [Accepted: 08/16/2023] [Indexed: 09/03/2023]
Abstract
OBJECTIVE Working memory (WM) deficits have frequently been linked to attention deficit hyperactivity disorder (ADHD). Despite previous studies suggested its high heritability, its genetic basis, especially in ADHD, remains unclear. The current study aimed to comprehensively explore the genetic basis of visual-spatial working memory (VSWM) in ADHD using wide-ranging genetic analyses. METHODS The current study recruited a cohort consisted of 802 ADHD individuals, all met DSM-IV ADHD diagnostic criteria. VSWM was assessed by Rey-Osterrieth complex figure test (RCFT), which is a widely used psychological test include four memory indexes: detail delayed (DD), structure delayed (SD), structure immediate (SI), detail immediate (DI). Genetic analyses were conducted at the single nucleotide polymorphism (SNP), gene, pathway, polygenic and protein network levels. Polygenic Risk Scores (PRS) were based on summary statistics of various psychiatric disorders, including ADHD, autism spectrum disorder (ASD), major depressive disorder (MDD), schizophrenia (SCZ), obsessive compulsive disorders (OCD), and substance use disorder (SUD). RESULTS Analyses at the single-marker level did not yield significant results (5E-08). However, the potential signals with P values less than E-05 and their mapped genes suggested the regulation of VSWM involved both ocular and neural system related genes, moreover, ADHD-related genes were also involved. The gene-based analysis found RAB11FIP1, whose encoded protein modulates several neurodevelopment processes and visual system, as significantly associated with DD scores (P = 1.96E-06, Padj = 0.036). Candidate pathway enrichment analyses (N = 53) found that forebrain neuron fate commitment significantly enriched in DD (P = 4.78E-04, Padj = 0.025), and dopamine transport enriched in SD (P = 5.90E-04, Padj = 0.031). We also observed a significant negative relationship between DD scores and ADHD PRS scores (P = 0.0025, Empirical P = 0.048). CONCLUSIONS Our results emphasized the joint contribution of ocular and neural genes in regulating VSWM. The study reveals a shared genetic basis between ADHD and VSWM, with GWAS indicating the involvement of ADHD-related genes in VSWM. Additionally, the PRS analysis identifies a significant relationship between ADHD-PRS and DD scores. Overall, our findings shed light on the genetic basis of VSWM deficits in ADHD, and may have important implications for future research and clinical practice.
Collapse
Affiliation(s)
- Yilu Zhao
- Peking University Sixth Hospital, Peking University Institute of Mental Health, National Clinical Research Center for Mental Disorders, Peking University Sixth Hospital), NHC Key Laboratory of Mental Health (Peking University), 51 Huayuan Bei Road, Beijing, 100191, China
| | - Yuanxin Zhong
- Peking University Sixth Hospital, Peking University Institute of Mental Health, National Clinical Research Center for Mental Disorders, Peking University Sixth Hospital), NHC Key Laboratory of Mental Health (Peking University), 51 Huayuan Bei Road, Beijing, 100191, China
- Department of Psychiatry, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, SAR, China
| | - Wei Chen
- Peking University Sixth Hospital, Peking University Institute of Mental Health, National Clinical Research Center for Mental Disorders, Peking University Sixth Hospital), NHC Key Laboratory of Mental Health (Peking University), 51 Huayuan Bei Road, Beijing, 100191, China
- Sichuan Provincial Center for Mental Health, The Center of Psychosomatic Medicine of Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
| | - Suhua Chang
- Peking University Sixth Hospital, Peking University Institute of Mental Health, National Clinical Research Center for Mental Disorders, Peking University Sixth Hospital), NHC Key Laboratory of Mental Health (Peking University), 51 Huayuan Bei Road, Beijing, 100191, China
| | - Qingjiu Cao
- Peking University Sixth Hospital, Peking University Institute of Mental Health, National Clinical Research Center for Mental Disorders, Peking University Sixth Hospital), NHC Key Laboratory of Mental Health (Peking University), 51 Huayuan Bei Road, Beijing, 100191, China
| | - Yufeng Wang
- Peking University Sixth Hospital, Peking University Institute of Mental Health, National Clinical Research Center for Mental Disorders, Peking University Sixth Hospital), NHC Key Laboratory of Mental Health (Peking University), 51 Huayuan Bei Road, Beijing, 100191, China
| | - Li Yang
- Peking University Sixth Hospital, Peking University Institute of Mental Health, National Clinical Research Center for Mental Disorders, Peking University Sixth Hospital), NHC Key Laboratory of Mental Health (Peking University), 51 Huayuan Bei Road, Beijing, 100191, China.
| |
Collapse
|
10
|
Hematologic Risk Factors for the Development of Retinopathy of Prematurity—A Retrospective Study. CHILDREN 2023; 10:children10030567. [PMID: 36980125 PMCID: PMC10047256 DOI: 10.3390/children10030567] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Revised: 03/12/2023] [Accepted: 03/16/2023] [Indexed: 03/19/2023]
Abstract
(1) Background: Retinopathy of prematurity (ROP) can cause severe visual impairment or even blindness. We aimed to assess the hematological risk factors that are associated with different stages of ROP in a cohort of preterm newborns, and to compare the clinical characteristics and therapeutic interventions between groups. (2) Methods: This retrospective study included 149 preterm newborns from a tertiary maternity hospital in Romania between January 2018 and December 2018, who were segregated into: Group 1 (with ROP, n = 59 patients), and Group 2 (without ROP, n = 90 patients). The patients that were affected by ROP were subsequently divided into the following subgroups: Subgroup 1 (Stage 1, n = 21), Subgroup 2 (Stage 2, n = 35), and Subgroup 3 (Stage 3, n = 25). The associations were analyzed using multivariate logistic regression and sensitivity analysis. (3) Results: Platelet mass indexes (PMI) that were determined in the first, seventh, and tenth days of life were significantly associated with Stage 1 ROP. PMI determined in the first day of life was also significantly associated with Stage 2 ROP. The sensitivity and specificity of these parameters were modest, ranging from 44 to 57%, and 59 to 63%. (4) Conclusions: PMI has a modest ability to predict the development of ROP.
Collapse
|
11
|
Sonny S, Yuan H, Chen S, Duncan MR, Chen P, Benny M, Young K, Park KK, Schmidt AF, Wu S. GSDMD deficiency ameliorates hyperoxia-induced BPD and ROP in neonatal mice. Sci Rep 2023; 13:143. [PMID: 36599874 DOI: 10.1038/s41598-022-27201-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Accepted: 12/28/2022] [Indexed: 01/06/2023] Open
Abstract
Bronchopulmonary dysplasia (BPD) and retinopathy of prematurity (ROP) are among the most common morbidities affecting extremely premature infants who receive oxygen therapy. Many clinical studies indicate that BPD is associated with advanced ROP. However, the mechanistic link between hyperoxia, BPD, and ROP remains to be explored. Gasdermin D (GSDMD) is a key executor of inflammasome-induced pyroptosis and inflammation. Inhibition of GSDMD has been shown to attenuate hyperoxia-induced BPD and brain injury in neonatal mice. The objective of this study was to further define the mechanistic roles of GSDMD in the pathogenesis of hyperoxia-induced BPD and ROP in mouse models. Here we show that global GSDMD knockout (GSDMD-KO) protects against hyperoxia-induced BPD by reducing macrophage infiltration, improving alveolarization and vascular development, and decreasing cell death. In addition, GSDMD deficiency prevented hyperoxia-induced ROP by reducing vasoobliteration and neovascularization, improving thinning of multiple retinal tissue layers, and decreasing microglial activation. RNA sequencing analyses of lungs and retinas showed that similar genes, including those from inflammatory, cell death, tissue remodeling, and tissue and vascular developmental signaling pathways, were induced by hyperoxia and impacted by GSDMD-KO in both models. These data highlight the importance of GSDMD in the pathogenesis of BPD and ROP and suggest that targeting GSDMD may be beneficial in preventing and treating BPD and ROP in premature infants.
Collapse
Affiliation(s)
- Sarah Sonny
- Neonatology and Batchelor Children Research Institute, University of Miami Miller School of Medicine, 1580 NW 10thAve, Miami, FL, 33136, USA
| | - Huijun Yuan
- Neonatology and Batchelor Children Research Institute, University of Miami Miller School of Medicine, 1580 NW 10thAve, Miami, FL, 33136, USA
| | - Shaoyi Chen
- Neonatology and Batchelor Children Research Institute, University of Miami Miller School of Medicine, 1580 NW 10thAve, Miami, FL, 33136, USA
| | - Matthew R Duncan
- Neonatology and Batchelor Children Research Institute, University of Miami Miller School of Medicine, 1580 NW 10thAve, Miami, FL, 33136, USA
| | - Pingping Chen
- Neonatology and Batchelor Children Research Institute, University of Miami Miller School of Medicine, 1580 NW 10thAve, Miami, FL, 33136, USA
| | - Merline Benny
- Neonatology and Batchelor Children Research Institute, University of Miami Miller School of Medicine, 1580 NW 10thAve, Miami, FL, 33136, USA
| | - Karen Young
- Neonatology and Batchelor Children Research Institute, University of Miami Miller School of Medicine, 1580 NW 10thAve, Miami, FL, 33136, USA
| | - Kevin K Park
- Department of Neurological Surgery, The Miami Project to Cure Paralysis, Miami, FL, USA
| | - Augusto F Schmidt
- Neonatology and Batchelor Children Research Institute, University of Miami Miller School of Medicine, 1580 NW 10thAve, Miami, FL, 33136, USA
| | - Shu Wu
- Neonatology and Batchelor Children Research Institute, University of Miami Miller School of Medicine, 1580 NW 10thAve, Miami, FL, 33136, USA.
| |
Collapse
|
12
|
Bujoreanu Bezman L, Tiutiuca C, Totolici G, Carneciu N, Bujoreanu FC, Ciortea DA, Niculet E, Fulga A, Alexandru AM, Stan DJ, Nechita A. Latest Trends in Retinopathy of Prematurity: Research on Risk Factors, Diagnostic Methods and Therapies. Int J Gen Med 2023; 16:937-949. [PMID: 36942030 PMCID: PMC10024537 DOI: 10.2147/ijgm.s401122] [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: 12/12/2022] [Accepted: 02/17/2023] [Indexed: 03/15/2023] Open
Abstract
Retinopathy of prematurity (ROP) is a vasoproliferative disorder with an imminent risk of blindness, in cases where early diagnosis and treatment are not performed. The doctors' constant motivation to give these fragile beings a chance at life with optimal visual acuity has never stopped, since Terry first described this condition. Thus, throughout time, several specific advancements have been made in the management of ROP. Apart from the most known risk factors, this narrative review brings to light the latest research about new potential risk factors, such as: proteinuria, insulin-like growth factor 1 (IGF-1) and blood transfusions. Digital imaging has revolutionized the management of retinal pathologies, and it is more and more used in identifying and staging ROP, particularly in the disadvantaged regions by the means of telescreening. Moreover, optical coherence tomography (OCT) and automated diagnostic tools based on deep learning offer new perspectives on the ROP diagnosis. The new therapeutical trend based on the use of anti-VEGF agents is increasingly used in the treatment of ROP patients, and recent research sustains the theory according to which these agents do not interfere with the neurodevelopment of premature babies.
Collapse
Affiliation(s)
- Laura Bujoreanu Bezman
- Department of Ophthalmology, “Sfantul Apostol Andrei” Emergency Clinical Hospital, Galati, Romania
- Department of Morphological and Functional Sciences, Faculty of Medicine and Pharmacy, “Dunărea de Jos” University, Galati, Romania
| | - Carmen Tiutiuca
- Department of Ophthalmology, “Sfantul Apostol Andrei” Emergency Clinical Hospital, Galati, Romania
- Clinical Surgical Department, Faculty of Medicine and Pharmacy, “Dunărea de Jos” University, Galati, Romania
- Correspondence: Carmen Tiutiuca, Clinical Surgical Department, Faculty of Medicine and Pharmacy, “Dunărea de Jos” University, Galati, 800008, Romania, Tel +40741330788, Email
| | - Geanina Totolici
- Department of Ophthalmology, “Sfantul Apostol Andrei” Emergency Clinical Hospital, Galati, Romania
- Clinical Surgical Department, Faculty of Medicine and Pharmacy, “Dunărea de Jos” University, Galati, Romania
| | - Nicoleta Carneciu
- Department of Ophthalmology, “Sfantul Apostol Andrei” Emergency Clinical Hospital, Galati, Romania
- Department of Morphological and Functional Sciences, Faculty of Medicine and Pharmacy, “Dunărea de Jos” University, Galati, Romania
| | - Florin Ciprian Bujoreanu
- Doctoral School of Biomedical Sciences, Faculty of Medicine and Pharmacy, “Dunărea de Jos” University, Galati, Romania
- Florin Ciprian Bujoreanu, Doctoral School of Biomedical Sciences, Faculty of Medicine and Pharmacy, “Dunărea de Jos” University, Galati, 800008, Romania, Tel +40741395844, Email
| | - Diana Andreea Ciortea
- Department of Pediatrics, “Sfantul Ioan” Emergency Clinical Hospital for Children, Galati, Romania
- Clinical Medical Department, Faculty of Medicine and Pharmacy, “Dunărea de Jos” University, Galati, Romania
| | - Elena Niculet
- Department of Morphological and Functional Sciences, Faculty of Medicine and Pharmacy, “Dunărea de Jos” University, Galati, Romania
- Doctoral School of Biomedical Sciences, Faculty of Medicine and Pharmacy, “Dunărea de Jos” University, Galati, Romania
| | - Ana Fulga
- Clinical Surgical Department, Faculty of Medicine and Pharmacy, “Dunărea de Jos” University, Galati, Romania
- Doctoral School of Biomedical Sciences, Faculty of Medicine and Pharmacy, “Dunărea de Jos” University, Galati, Romania
| | - Anamaria Madalina Alexandru
- Doctoral School of Biomedical Sciences, Faculty of Medicine and Pharmacy, “Dunărea de Jos” University, Galati, Romania
- Department of Neonatology, “Sfantul Apostol Andrei” Emergency Clinical Hospital, Galati, Romania
| | - Daniela Jicman Stan
- Doctoral School of Biomedical Sciences, Faculty of Medicine and Pharmacy, “Dunărea de Jos” University, Galati, Romania
| | - Aurel Nechita
- Department of Pediatrics, “Sfantul Ioan” Emergency Clinical Hospital for Children, Galati, Romania
- Clinical Medical Department, Faculty of Medicine and Pharmacy, “Dunărea de Jos” University, Galati, Romania
| |
Collapse
|
13
|
Dammann O, Hartnett ME, Stahl A. Retinopathy of prematurity. Dev Med Child Neurol 2022; 65:625-631. [PMID: 36408783 DOI: 10.1111/dmcn.15468] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Revised: 10/12/2022] [Accepted: 10/17/2022] [Indexed: 11/22/2022]
Abstract
Retinopathy of prematurity (ROP) is a devastating neurovascular disease of the retina in newborn infants that can lead to vision deficits or even blindness. In this concise review we discuss our current knowledge about diagnosis, etiology, pathogenesis, intervention, and outcomes of the disease. Major advancements have been made both in categorizing the disease in the new International Classification of Retinopathy of Prematurity, Third Edition classification and in treating severe ROP with anti-vascular endothelial growth factor (VEGF) agents. New development always creates new questions and opens up new areas of research. We will discuss in this review both the benefits and downsides of the new anti-VEGF treatment approaches in ROP, especially in light of our improved understanding of the underlying ROP pathophysiology. We also offer pointers to areas where more research is needed.
Collapse
Affiliation(s)
- Olaf Dammann
- Tufts University School of Medicine, Department of Public Health & Community Medicine, MA, Boston, USA.,Department of Obstetrics & Gynecology, Hannover Medical School, Hannover, Germany.,Department of Neuromedicine and Movement Science, Norwegian University of Science & Technology, Trondheim, Norway
| | - M Elizabeth Hartnett
- John A Moran Eye Center, Department of Ophthalmology and Visual Sciences, University of Utah Health, UT, Salt Lake City, USA
| | - Andreas Stahl
- Department of Ophthalmology Greifswald, University Medicine Greifswald, Mecklenburg-Vorpommern, Germany
| |
Collapse
|
14
|
Kim H, Kim J, Ryu J. Noncoding RNAs as a novel approach to target retinopathy of prematurity. Front Pharmacol 2022; 13:1033341. [PMID: 36386230 PMCID: PMC9641647 DOI: 10.3389/fphar.2022.1033341] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Accepted: 10/11/2022] [Indexed: 06/21/2024] Open
Abstract
Retinopathy of prematurity (ROP), a vascular disease characterized by abnormal vessel development in the retina, has become a primary cause of blindness in children around the world. ROP can be developed during two different phases: vessel loss and vessel proliferation. Once preterm infants with immature retinal vessel growth are exposed to high level of oxygen inside the incubator, vessel loss can occur. When infants are exposed to room air, they may experience the proliferation of vessels in the retina. Although multiple factors are reported to be involved in the pathogenesis of ROP, including vaso-endothelial growth factors (VEGFs) and hypoxia-inducible factors, the pathogenesis of ROP is not completely understood. Although laser therapy and pharmacologic agents, such as anti-VEGF agents, have been commonly used to treat ROP, the incidence of ROP is rapidly rising. Given that current therapies can be invasive and long-term effects are not fully known, the search for novel therapeutic targets with less destructive properties needs to be considered. Within the last decade, the field of noncoding RNA therapy has shown potential as next-generation therapy to treat diverse diseases. In this review, we introduce various noncoding RNAs regulating ROP and discuss their role as potential therapeutic targets in ROP.
Collapse
Affiliation(s)
- Hyunjong Kim
- Vessel-Organ Interaction Research Center, College of Pharmacy, Kyungpook National University, Daegu, South Korea
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Kyungpook National University, Daegu, South Korea
| | - Jaesub Kim
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Kyungpook National University, Daegu, South Korea
| | - Juhee Ryu
- Vessel-Organ Interaction Research Center, College of Pharmacy, Kyungpook National University, Daegu, South Korea
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Kyungpook National University, Daegu, South Korea
| |
Collapse
|
15
|
Sanchez M, Hamel D, Bajon E, Duhamel F, Bhosle VK, Zhu T, Rivera JC, Dabouz R, Nadeau-Vallée M, Sitaras N, Tremblay DÉ, Omri S, Habelrih T, Rouget R, Hou X, Gobeil F, Joyal JS, Sapieha P, Mitchell G, Ribeiro-Da-Silva A, Mohammad Nezhady MA, Chemtob S. The Succinate Receptor SUCNR1 Resides at the Endoplasmic Reticulum and Relocates to the Plasma Membrane in Hypoxic Conditions. Cells 2022; 11:2185. [PMID: 35883628 PMCID: PMC9321536 DOI: 10.3390/cells11142185] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Revised: 07/08/2022] [Accepted: 07/09/2022] [Indexed: 12/24/2022] Open
Abstract
The GPCR SUCNR1/GPR91 exerts proangiogenesis upon stimulation with the Krebs cycle metabolite succinate. GPCR signaling depends on the surrounding environment and intracellular localization through location bias. Here, we show by microscopy and by cell fractionation that in neurons, SUCNR1 resides at the endoplasmic reticulum (ER), while being fully functional, as shown by calcium release and the induction of the expression of the proangiogenic gene for VEGFA. ER localization was found to depend upon N-glycosylation, particularly at position N8; the nonglycosylated mutant receptor localizes at the plasma membrane shuttled by RAB11. This SUCNR1 glycosylation is physiologically regulated, so that during hypoxic conditions, SUCNR1 is deglycosylated and relocates to the plasma membrane. Downstream signal transduction of SUCNR1 was found to activate the prostaglandin synthesis pathway through direct interaction with COX-2 at the ER; pharmacologic antagonism of the PGE2 EP4 receptor (localized at the nucleus) was found to prevent VEGFA expression. Concordantly, restoring the expression of SUCNR1 in the retina of SUCNR1-null mice renormalized vascularization; this effect is markedly diminished after transfection of the plasma membrane-localized SUCNR1 N8A mutant, emphasizing that ER localization of the succinate receptor is necessary for proper vascularization. These findings uncover an unprecedented physiologic process where GPCR resides at the ER for signaling function.
Collapse
Affiliation(s)
- Melanie Sanchez
- Department of Pharmacology and Therapeutics, McGill University, Montréal, QC H3A 1A3, Canada; (M.S.); (V.K.B.); (R.D.); (R.R.); (A.R.-D.-S.)
| | - David Hamel
- Department of Pharmacology, Université de Montréal, Montréal, QC H3C 3J7, Canada; (D.H.); (F.D.); (M.N.-V.); (N.S.); (D.-É.T.); (T.H.)
| | - Emmanuel Bajon
- Department of Pediatrics, Research Center-CHU Ste-Justine, Montréal, QC H3T 1C5, Canada; (E.B.); (T.Z.); (X.H.); (J.-S.J.); (G.M.)
| | - François Duhamel
- Department of Pharmacology, Université de Montréal, Montréal, QC H3C 3J7, Canada; (D.H.); (F.D.); (M.N.-V.); (N.S.); (D.-É.T.); (T.H.)
| | - Vikrant K. Bhosle
- Department of Pharmacology and Therapeutics, McGill University, Montréal, QC H3A 1A3, Canada; (M.S.); (V.K.B.); (R.D.); (R.R.); (A.R.-D.-S.)
- Cell Biology Program, The Hospital for Sick Children Research Institute, Toronto, ON M5G 1X8, Canada
| | - Tang Zhu
- Department of Pediatrics, Research Center-CHU Ste-Justine, Montréal, QC H3T 1C5, Canada; (E.B.); (T.Z.); (X.H.); (J.-S.J.); (G.M.)
| | - Jose Carlos Rivera
- Department of Ophthalmology, Research Center of Hôpital Maisonneuve-Rosemont, Université de Montréal, Montréal, QC H1T 2M4, Canada; (J.C.R.); (S.O.); (P.S.)
| | - Rabah Dabouz
- Department of Pharmacology and Therapeutics, McGill University, Montréal, QC H3A 1A3, Canada; (M.S.); (V.K.B.); (R.D.); (R.R.); (A.R.-D.-S.)
| | - Mathieu Nadeau-Vallée
- Department of Pharmacology, Université de Montréal, Montréal, QC H3C 3J7, Canada; (D.H.); (F.D.); (M.N.-V.); (N.S.); (D.-É.T.); (T.H.)
| | - Nicholas Sitaras
- Department of Pharmacology, Université de Montréal, Montréal, QC H3C 3J7, Canada; (D.H.); (F.D.); (M.N.-V.); (N.S.); (D.-É.T.); (T.H.)
| | - David-Étienne Tremblay
- Department of Pharmacology, Université de Montréal, Montréal, QC H3C 3J7, Canada; (D.H.); (F.D.); (M.N.-V.); (N.S.); (D.-É.T.); (T.H.)
| | - Samy Omri
- Department of Ophthalmology, Research Center of Hôpital Maisonneuve-Rosemont, Université de Montréal, Montréal, QC H1T 2M4, Canada; (J.C.R.); (S.O.); (P.S.)
| | - Tiffany Habelrih
- Department of Pharmacology, Université de Montréal, Montréal, QC H3C 3J7, Canada; (D.H.); (F.D.); (M.N.-V.); (N.S.); (D.-É.T.); (T.H.)
| | - Raphael Rouget
- Department of Pharmacology and Therapeutics, McGill University, Montréal, QC H3A 1A3, Canada; (M.S.); (V.K.B.); (R.D.); (R.R.); (A.R.-D.-S.)
| | - Xin Hou
- Department of Pediatrics, Research Center-CHU Ste-Justine, Montréal, QC H3T 1C5, Canada; (E.B.); (T.Z.); (X.H.); (J.-S.J.); (G.M.)
| | - Fernand Gobeil
- Department of Pharmacology, Université de Sherbrooke, Sherbrooke, QC J1H 5N4, Canada;
| | - Jean-Sébastien Joyal
- Department of Pediatrics, Research Center-CHU Ste-Justine, Montréal, QC H3T 1C5, Canada; (E.B.); (T.Z.); (X.H.); (J.-S.J.); (G.M.)
- Department of Ophthalmology, Research Center of Hôpital Maisonneuve-Rosemont, Université de Montréal, Montréal, QC H1T 2M4, Canada; (J.C.R.); (S.O.); (P.S.)
| | - Przemyslaw Sapieha
- Department of Ophthalmology, Research Center of Hôpital Maisonneuve-Rosemont, Université de Montréal, Montréal, QC H1T 2M4, Canada; (J.C.R.); (S.O.); (P.S.)
| | - Grant Mitchell
- Department of Pediatrics, Research Center-CHU Ste-Justine, Montréal, QC H3T 1C5, Canada; (E.B.); (T.Z.); (X.H.); (J.-S.J.); (G.M.)
| | - Alfredo Ribeiro-Da-Silva
- Department of Pharmacology and Therapeutics, McGill University, Montréal, QC H3A 1A3, Canada; (M.S.); (V.K.B.); (R.D.); (R.R.); (A.R.-D.-S.)
| | - Mohammad Ali Mohammad Nezhady
- Department of Pediatrics, Research Center-CHU Ste-Justine, Montréal, QC H3T 1C5, Canada; (E.B.); (T.Z.); (X.H.); (J.-S.J.); (G.M.)
- Program of Molecular Biology, Faculty of Medicine, Université de Montréal, Montréal, QC H3T 1J4, Canada
- Research Center-CHU Ste-Justine, Departments of Pediatrics, Ophthalmology, and Pharmacology, Faculty of Medicine, Université de Montréal, 3175, Chemin Côte Ste-Catherine, Montréal, QC H3T 1C5, Canada
| | - Sylvain Chemtob
- Department of Pharmacology and Therapeutics, McGill University, Montréal, QC H3A 1A3, Canada; (M.S.); (V.K.B.); (R.D.); (R.R.); (A.R.-D.-S.)
- Department of Pharmacology, Université de Montréal, Montréal, QC H3C 3J7, Canada; (D.H.); (F.D.); (M.N.-V.); (N.S.); (D.-É.T.); (T.H.)
- Department of Pediatrics, Research Center-CHU Ste-Justine, Montréal, QC H3T 1C5, Canada; (E.B.); (T.Z.); (X.H.); (J.-S.J.); (G.M.)
- Department of Ophthalmology, Research Center of Hôpital Maisonneuve-Rosemont, Université de Montréal, Montréal, QC H1T 2M4, Canada; (J.C.R.); (S.O.); (P.S.)
- Research Center-CHU Ste-Justine, Departments of Pediatrics, Ophthalmology, and Pharmacology, Faculty of Medicine, Université de Montréal, 3175, Chemin Côte Ste-Catherine, Montréal, QC H3T 1C5, Canada
| |
Collapse
|
16
|
Zhao C, Liu Y, Meng J, Wang X, Liu X, Li W, Zhou Q, Xiang J, Li N, Hou S. LGALS3BP in Microglia Promotes Retinal Angiogenesis Through PI3K/AKT Pathway During Hypoxia. Invest Ophthalmol Vis Sci 2022; 63:25. [PMID: 35895036 PMCID: PMC9344220 DOI: 10.1167/iovs.63.8.25] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Purpose Retinal microglia promote angiogenesis and vasculopathy in oxygen-induced retinopathy (OIR); however, its specific molecular mechanism in the formation of retinal angiogenesis remains unclear. The lectin galactoside-binding soluble 3 binding protein (LGALS3BP), a member of the scavenger receptor cysteine-rich (SRCR) domain protein family, is involved in tumor neovascularization, and we therefore hypothesized that LGALS3BP plays an active role in microglia-induced angiogenesis. Methods The expression of LGALS3BP in microglia was detected by immunofluorescence, RT-qPCR, and western blotting. Functional assays of human umbilical vein endothelial cells (HUVECs) such as migration, proliferation, and tube formation were measured by Transwell, EdU, and Matrigel assays. Angiogenesis-related factors and PI3K/AKT levels were detected by western blotting. The relationship between LGALS3BP and PI3K or HIF-1α was investigated by immunoprecipitation. Results Our results showed that the expression of LGALS3BP was significantly increased in microglia surrounding neovascularization of the OIR mice and was also upregulated in human microglial clone 3 (HMC3) cells after hypoxia. Moreover, HUVECs co-cultured with hypoxic HMC3 cells showed increased migration, proliferation, and tube formation, as well as levels of angiogenesis-related factor. However, the proangiogenic ability and angiogenesis-related factor expression of HMC3 cells was suppressed after silencing LGALS3BP. LGALS3BP induces the upregulation of angiogenesis-related factors through the PI3K/AKT pathway and then promotes angiogenesis in microglia. Conclusions Collectively, our findings suggest that LGALS3BP in microglia plays an important role in angiogenesis, suggesting a potential therapeutic target of LGALS3BP for angiogenesis.
Collapse
Affiliation(s)
- Chenyang Zhao
- The First Affiliated Hospital of Chongqing Medical University, Chongqing, China.,Chongqing Key Laboratory of Ophthalmology, Chongqing, China.,Chongqing Branch of National Clinical Research Center for Ocular Diseases, Chongqing, China
| | - Yusen Liu
- The First Affiliated Hospital of Chongqing Medical University, Chongqing, China.,Chongqing Key Laboratory of Ophthalmology, Chongqing, China.,Chongqing Branch of National Clinical Research Center for Ocular Diseases, Chongqing, China
| | - Jiayu Meng
- The First Affiliated Hospital of Chongqing Medical University, Chongqing, China.,Chongqing Key Laboratory of Ophthalmology, Chongqing, China.,Chongqing Branch of National Clinical Research Center for Ocular Diseases, Chongqing, China
| | - Xiaotang Wang
- The First Affiliated Hospital of Chongqing Medical University, Chongqing, China.,Chongqing Key Laboratory of Ophthalmology, Chongqing, China.,Chongqing Branch of National Clinical Research Center for Ocular Diseases, Chongqing, China
| | - Xianyang Liu
- The First Affiliated Hospital of Chongqing Medical University, Chongqing, China.,Chongqing Key Laboratory of Ophthalmology, Chongqing, China.,Chongqing Branch of National Clinical Research Center for Ocular Diseases, Chongqing, China
| | - Wanqian Li
- The First Affiliated Hospital of Chongqing Medical University, Chongqing, China.,Chongqing Key Laboratory of Ophthalmology, Chongqing, China.,Chongqing Branch of National Clinical Research Center for Ocular Diseases, Chongqing, China
| | - Qian Zhou
- The First Affiliated Hospital of Chongqing Medical University, Chongqing, China.,Chongqing Key Laboratory of Ophthalmology, Chongqing, China.,Chongqing Branch of National Clinical Research Center for Ocular Diseases, Chongqing, China
| | - Junjie Xiang
- The First Affiliated Hospital of Chongqing Medical University, Chongqing, China.,Chongqing Key Laboratory of Ophthalmology, Chongqing, China.,Chongqing Branch of National Clinical Research Center for Ocular Diseases, Chongqing, China
| | - Na Li
- College of Basic Medicine, Chongqing Medical University, Chongqing, China
| | - Shengping Hou
- The First Affiliated Hospital of Chongqing Medical University, Chongqing, China.,Chongqing Key Laboratory of Ophthalmology, Chongqing, China.,Chongqing Branch of National Clinical Research Center for Ocular Diseases, Chongqing, China
| |
Collapse
|
17
|
RNA-Seq Provides Insights into VEGF-Induced Signaling in Human Retinal Microvascular Endothelial Cells: Implications in Retinopathy of Prematurity. Int J Mol Sci 2022; 23:ijms23137354. [PMID: 35806359 PMCID: PMC9266443 DOI: 10.3390/ijms23137354] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Revised: 06/19/2022] [Accepted: 06/28/2022] [Indexed: 02/04/2023] Open
Abstract
The pathophysiology of retinopathy of prematurity (ROP) is postulated to first involve delayed intraretinal vascularization, followed by intravitreal neovascularization (IVNV). Although intravitreal agents that reduce the bioactivity of vascular endothelial growth factor (VEGF) are used to treat IVNV, concerns exist regarding their effects on intraretinal vascularization. In an experimental ROP model, VEGF receptor 2 (VEGFR2) knockdown in retinal endothelial cells reduced IVNV and promoted intraretinal vascularization, whereas knockdown of a downstream effector, signal transducer and activator of transcription 3 (STAT3) in retinal endothelial cells only reduced IVNV. In this study, we tested the hypothesis that the different pathways involved in VEGF-triggered VEGFR2 signaling and VEGF-triggered STAT3 signaling in retinal endothelial cells would allow us to delineate signaling pathways involved in IVNV from those involved in intraretinal vascularization in ROP. To address our hypothesis, we used RNA-sequencing and pathway enrichment analysis to determine changes in the transcriptome of cultured human retinal microvascular endothelial cells (HRMECs). Of the enriched pathways, inactivation of oncostatin M signaling was predicted by either KDR or STAT3 knockdown in the presence of VEGF. Activation of kinetochore metaphase signaling was predicted by KDR knockdown, whereas inactivation was predicted by STAT3 knockdown in the presence of VEGF. Inactivation of signaling by the Rho family of GTPases was predicted by KDR knockdown, but activation was predicted by STAT3 knockdown in the presence of VEGF. Taken together, our data identified unique signaling pathway differences between VEGF-triggered VEGFR2 and VEGF-triggered STAT3 in HRMECs that might have implications in ROP.
Collapse
|