1
|
Bai Y, Wang R, Jin C, Wang L, Tang Y, Wang H, Ding K, Cai S. Impacts of sulfate polysaccharide JCS1S2 on retinal neovascularization in oxygen-induced retinopathy rats. Front Pharmacol 2025; 16:1499420. [PMID: 40271076 PMCID: PMC12014746 DOI: 10.3389/fphar.2025.1499420] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2024] [Accepted: 03/19/2025] [Indexed: 04/25/2025] Open
Abstract
Retinal neovascularization, a pathological form of angiogenesis, is a leading cause of blindness. This study investigated the effects of Dendrobium sulfate polysaccharide JCS1S2, derived from Dendrobium chrysogenum, on oxygen-induced retinopathy (OIR) in rat models and Müller cells of the rat retina. To this end, we established an OIR rat model and divided the rats into three primary groups, namely, Group I (control), Group II (OIR), and Group III (OIR + JCS1S2). Group III was further subdivided into three subgroups treated with different concentrations of JCS1S2 (10 μg/μL, 20 μg/μL, and 40 μg/μL). After finding the optimal concentration of JCS1S2 by ADP and HE, PCR, Western blot and transcriptome sequencing were used to analyze the role of JCS1S2 in Müller cells of OIR rats and rat retinas. ADP and hematoxylin and eosin (HE) staining revealed that JCS1S2 dose-dependently inhibited retinal neovascularization. Quantitative polymerase chain reaction (qPCR) and Western blot analyses showed significant downregulation of vascular endothelial growth factor A (VEGF-A), VEGF-B, VEGF-D, VEGF receptor 1 (VEGF-R1), and VEGF receptor 2 (VEGF-R2) following JCS1S2 treatment. Transcriptome analysis suggested that JCS1S2 may suppress the activation of the Toll-like receptor (TLR) signaling pathway, regulate the expression of genes associated with endothelial activation and angiogenesis, and participate in the inflammatory and metabolic pathways of the retina. Western blotting data indicate that JCS1S2 can markedly reduce abnormal retinal angiogenesis and Müller cell activation in OIR rats through the TLR4/p-NF-κB/VEGF pathway, JCS1S2 may have the potential as a therapeutic agent for retinal neovascularization.
Collapse
Affiliation(s)
- Yang Bai
- Department of Ophthalmology, Special Key Laboratory of Ocular Diseases of Guizhou Province, Affiliated Hospital of Zunyi Medical University, Guizhou Eye Hospital, Guizhou Provincial Branch of National Eye Disease Clinical Research Center, Zunyi Medical University, Zunyi, Guizhou Province, China
| | - Rui Wang
- Department of Ophthalmology, Special Key Laboratory of Ocular Diseases of Guizhou Province, Affiliated Hospital of Zunyi Medical University, Guizhou Eye Hospital, Guizhou Provincial Branch of National Eye Disease Clinical Research Center, Zunyi Medical University, Zunyi, Guizhou Province, China
| | - Can Jin
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Science, Zhongshan, China
- Glycochemistry and Glycobiology Lab, CAS Key Laboratory of Receptor Research, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Beijing, China
| | - Lili Wang
- Department of Ophthalmology, Special Key Laboratory of Ocular Diseases of Guizhou Province, Affiliated Hospital of Zunyi Medical University, Guizhou Eye Hospital, Guizhou Provincial Branch of National Eye Disease Clinical Research Center, Zunyi Medical University, Zunyi, Guizhou Province, China
| | - Yun Tang
- Department of Ophthalmology, Special Key Laboratory of Ocular Diseases of Guizhou Province, Affiliated Hospital of Zunyi Medical University, Guizhou Eye Hospital, Guizhou Provincial Branch of National Eye Disease Clinical Research Center, Zunyi Medical University, Zunyi, Guizhou Province, China
| | - Han Wang
- Department of Ophthalmology, Special Key Laboratory of Ocular Diseases of Guizhou Province, Affiliated Hospital of Zunyi Medical University, Guizhou Eye Hospital, Guizhou Provincial Branch of National Eye Disease Clinical Research Center, Zunyi Medical University, Zunyi, Guizhou Province, China
| | - Kan Ding
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Science, Zhongshan, China
- Glycochemistry and Glycobiology Lab, CAS Key Laboratory of Receptor Research, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Beijing, China
| | - Shanjun Cai
- Department of Ophthalmology, Special Key Laboratory of Ocular Diseases of Guizhou Province, Affiliated Hospital of Zunyi Medical University, Guizhou Eye Hospital, Guizhou Provincial Branch of National Eye Disease Clinical Research Center, Zunyi Medical University, Zunyi, Guizhou Province, China
| |
Collapse
|
2
|
Zhao J, Zhang M, Wang Y, He F, Zhang Q. Identification of cuproptosis-related genes in septic shock based on bioinformatic analysis. PLoS One 2024; 19:e0315219. [PMID: 39652607 PMCID: PMC11627398 DOI: 10.1371/journal.pone.0315219] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Accepted: 11/21/2024] [Indexed: 12/12/2024] Open
Abstract
BACKGROUND Septic shock is a life-threatening condition characterized by a failure of organ systems and a high mortality rate. Cuproptosis is a new form of cell death that is triggered by copper overload. However, the relationship between cuproptosis-related genes and septic shock remains unclear. METHODS The GSE26440 dataset from the GEO database was used to screen differentially expressed genes (DEGs) between control and septic shock samples. Additionally, hub genes related to the progression of septic shock and cuproptosis were screened by Venn analysis. RT-qPCR was utilized to validate the expression of hub genes in peripheral blood lymphocytes from septic shock patients and healthy controls. Next, functional analysis and immune cells infiltration were performed. RESULTS SLC31A1 and MTF1 levels were obviously elevated and LIAS and LIPT1 levels were downregulated in septic shock samples, compared to normal controls. The diagnostic values of the four genes were confirmed with receiver operating characteristic (ROC) curves. Additionally, SLC31A1 and MTF1 showed a positive correlation with natural killer cells and LIAS and LIPT1 exhibited a positive correlation with CD8+ T cells. Furthermore, compared to low-level groups, MAPK signaling was activated in the high-SLC31A1 level group, VEGF signaling was activated in the high-MTF1 level group and lipoic acid metabolism was activated in high-LIAS and high-LIPT1 level groups. CONCLUSION This study demonstrates that SLC31A1, MTF1, LIAS, and LIPT1 are dysregulated in septic shock samples, and these genes exhibit potential diagnostic efficacy in septic shock, suggesting that these genes may be potential biomarkers for the diagnosis of septic shock.
Collapse
Affiliation(s)
- Jintong Zhao
- Department of Critical Medicine, Zibo Central Hospital, Zibo, China
| | - Meng Zhang
- Department of Critical Medicine, Qingdao Central Hospital, Qingdao, China
| | - Ying Wang
- Department of Nosocomial Infection, Qingdao Cancer Hospital, Qingdao, China
| | - Feifei He
- Department of Critical Medicine, Qingdao Hiser Hospital, Affiliated Hospital of Qingdao University (Qingdao Traditional Chinese Medicine Hospital), Qingdao, China
| | - Qiang Zhang
- Department of Critical Medicine, Zibo Central Hospital, Zibo, China
| |
Collapse
|
3
|
Torres-Espin A, Radabaugh HL, Treiman S, Fitzsimons SS, Harvey D, Chou A, Lindbergh CA, Casaletto KB, Goldberger L, Staffaroni AM, Maillard P, Miller BL, DeCarli C, Hinman JD, Ferguson AR, Kramer JH, Elahi FM. Sexually dimorphic differences in angiogenesis markers are associated with brain aging trajectories in humans. Sci Transl Med 2024; 16:eadk3118. [PMID: 39602511 DOI: 10.1126/scitranslmed.adk3118] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Accepted: 11/06/2024] [Indexed: 11/29/2024]
Abstract
Aberrant angiogenesis could contribute to the development of cognitive impairment and represent a therapeutic target for preventing dementia. However, most studies addressing angiogenesis and cognitive impairment focus on model organisms. To test the relevance of angiogenesis to human cognitive aging, we evaluated associations of circulating blood markers of angiogenesis with brain aging trajectories in a pooled two-center sample from deeply phenotyped longitudinal human cohorts (n = 435; female = 207, age = 74 ± 9) using cognitive assessments, biospecimens, structural brain imaging, and clinical data. Blood markers included ligands involved in angiogenesis and vascular function such as basic fibroblast growth factor (bFGF), members of the vascular endothelial growth factor family (VEGFA, VEGFB, and VEGFC), and placental growth factor (PlGF), in addition to their receptors VEGF receptor 1 (VEGFR1) and tyrosine kinase with immunoglobulin and EGF homology domain 2 (Tie2). Machine learning and traditional statistics revealed sexually dimorphic associations of plasma angiogenic growth factors with brain aging outcomes, including executive function and gray matter atrophy. Specifically, markers of angiogenesis were associated with higher executive function and less brain atrophy in younger women (not men), a directionality of association that reversed around age 75. Higher concentrations of bFGF, known for pleiotropic effects on multiple cell types, predicted favorable cognitive trajectories in both women and men. An independent sample from a multicenter dataset (MarkVCID; n = 80; female = 30, age = 73 ± 9) was used to externally validate these findings. In conclusion, this analysis demonstrates the association of angiogenesis to human brain aging, with potential therapeutic implications for vascular cognitive impairment and dementia.
Collapse
Affiliation(s)
- Abel Torres-Espin
- School of Public Health Sciences, Faculty of Health, University of Waterloo, Waterloo, ON N2L 3G1, Canada
- Department of Neurological Surgery, Brain and Spinal Injury Center (BASIC), Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Hannah L Radabaugh
- Department of Neurological Surgery, Brain and Spinal Injury Center (BASIC), Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Scott Treiman
- Department of Neurological Surgery, Brain and Spinal Injury Center (BASIC), Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Stephen S Fitzsimons
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Danielle Harvey
- Department of Public Health Sciences, University of California, Davis, Davis, CA 95616, USA
| | - Austin Chou
- Department of Neurological Surgery, Brain and Spinal Injury Center (BASIC), Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Cutter A Lindbergh
- Department of Neurology, Memory and Aging Center, Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA 94158, USA
- Department of Psychiatry, University of Connecticut School of Medicine, Farmington, CT 06032, USA
| | - Kaitlin B Casaletto
- Department of Neurology, Memory and Aging Center, Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Lauren Goldberger
- Department of Neurology, Memory and Aging Center, Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Adam M Staffaroni
- Department of Neurology, Memory and Aging Center, Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Pauline Maillard
- Department of Neurology, University of California, Davis, Davis, CA 95817, USA
| | - Bruce L Miller
- Department of Neurology, Memory and Aging Center, Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Charles DeCarli
- Department of Neurology, University of California, Davis, Davis, CA 95817, USA
| | - Jason D Hinman
- Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Adam R Ferguson
- Department of Neurological Surgery, Brain and Spinal Injury Center (BASIC), Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA 94158, USA
- San Francisco Veterans Affairs Health Care System, San Francisco, CA 94121, USA
| | - Joel H Kramer
- Department of Neurology, Memory and Aging Center, Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Fanny M Elahi
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Department of Neurology, Memory and Aging Center, Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA 94158, USA
- James J. Peters Veterans Affairs Health Care System, Bronx, NY 10468, USA
| |
Collapse
|
4
|
Raja Xavier JP, Okumura T, Apweiler M, Chacko NA, Singh Y, Brucker SY, Takeda S, Lang F, Salker MS. Placental growth factor mediates pathological uterine angiogenesis by activating the NFAT5-SGK1 signaling axis in the endometrium: implications for preeclampsia development. Biol Res 2024; 57:55. [PMID: 39152497 PMCID: PMC11330076 DOI: 10.1186/s40659-024-00526-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2024] [Accepted: 06/26/2024] [Indexed: 08/19/2024] Open
Abstract
After menstruation the uterine spiral arteries are repaired through angiogenesis. This process is tightly regulated by the paracrine communication between endometrial stromal cells (EnSCs) and endothelial cells. Any molecular aberration in these processes can lead to complications in pregnancy including miscarriage or preeclampsia (PE). Placental growth factor (PlGF) is a known contributing factor for pathological angiogenesis but the mechanisms remain poorly understood. In this study, we investigated whether PlGF contributes to pathological uterine angiogenesis by disrupting EnSCs and endothelial paracrine communication. We observed that PlGF mediates a tonicity-independent activation of nuclear factor of activated T cells 5 (NFAT5) in EnSCs. NFAT5 activated downstream targets including SGK1, HIF-1α and VEGF-A. In depth characterization of PlGF - conditioned medium (CM) from EnSCs using mass spectrometry and ELISA methods revealed low VEGF-A and an abundance of extracellular matrix organization associated proteins. Secreted factors in PlGF-CM impeded normal angiogenic cues in endothelial cells (HUVECs) by downregulating Notch-VEGF signaling. Interestingly, PlGF-CM failed to support human placental (BeWo) cell invasion through HUVEC monolayer. Inhibition of SGK1 in EnSCs improved angiogenic effects in HUVECs and promoted BeWo invasion, revealing SGK1 as a key intermediate player modulating PlGF mediated anti-angiogenic signaling. Taken together, perturbed PlGF-NFAT5-SGK1 signaling in the endometrium can contribute to pathological uterine angiogenesis by negatively regulating EnSCs-endothelial crosstalk resulting in poor quality vessels in the uterine microenvironment. Taken together the signaling may impact on normal trophoblast invasion and thus placentation and, may be associated with an increased risk of complications such as PE.
Collapse
Affiliation(s)
- Janet P Raja Xavier
- Department of Women's Health, University of Tübingen, 72076, Calwerstraße 7/6, Tübingen, Germany
| | - Toshiyuki Okumura
- Department of Obstetrics and Gynaecology, Juntendo University School of Medicine, Tokyo, Japan
| | - Melina Apweiler
- Department of Women's Health, University of Tübingen, 72076, Calwerstraße 7/6, Tübingen, Germany
| | - Nirzari A Chacko
- Department of Women's Health, University of Tübingen, 72076, Calwerstraße 7/6, Tübingen, Germany
| | - Yogesh Singh
- Department of Women's Health, University of Tübingen, 72076, Calwerstraße 7/6, Tübingen, Germany
- Institute of Medical Genetics and Applied Genomics, University of Tübingen, Tübingen, Germany
| | - Sara Y Brucker
- Department of Women's Health, University of Tübingen, 72076, Calwerstraße 7/6, Tübingen, Germany
| | - Satoru Takeda
- Department of Obstetrics and Gynaecology, Juntendo University School of Medicine, Tokyo, Japan
| | - Florian Lang
- Department of Physiology, University of Tübingen, Tübingen, Germany
| | - Madhuri S Salker
- Department of Women's Health, University of Tübingen, 72076, Calwerstraße 7/6, Tübingen, Germany.
| |
Collapse
|
5
|
Ożóg MK, Nowak-Wąs M, Rokicki W. Pathophysiology and clinical aspects of epiretinal membrane - review. Front Med (Lausanne) 2023; 10:1121270. [PMID: 37636571 PMCID: PMC10447902 DOI: 10.3389/fmed.2023.1121270] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2022] [Accepted: 07/24/2023] [Indexed: 08/29/2023] Open
Abstract
The epiretinal membrane (ERM) is a pathological tissue formed at the vitreoretinal interface. The formation of this tissue is associated with numerous symptoms related to disturbances of vision. These types of lesions may arise idiopathically or be secondary to eye diseases, injuries and retinal surgeries. ERM tissue contains numerous cell types and numerous cytokines, which participate in its formation. The aim of this paper is to summarize information about the etiology, epidemiology, pathophysiology and treatment of ERM, with a brief description of the main cells that build the ERM - as well as the cytokines and molecules related to ERM pathogenesis - being provided in addition.
Collapse
Affiliation(s)
- Mateusz Kamil Ożóg
- Department of Histology and Cell Pathology, Faculty of Medical Sciences in Zabrze, Medical University of Silesia, Katowice, Poland
- Department of Histology, Cytophysiology and Embryology, Faculty of Medicine, Academy of Silesia, Zabrze, Poland
| | - Marta Nowak-Wąs
- Department of Histology and Cell Pathology, Faculty of Medical Sciences in Zabrze, Medical University of Silesia, Katowice, Poland
- Department of Ophthalmology, Kornel Gibiński University Clinical Center, Medical University of Silesia, Katowice, Poland
| | - Wojciech Rokicki
- Department of Ophthalmology, Kornel Gibiński University Clinical Center, Medical University of Silesia, Katowice, Poland
- Department of Ophthalmology, Faculty of Medical Sciences in Katowice, Medical University of Silesia, Katowice, Poland
| |
Collapse
|
6
|
Donato L, Scimone C, Alibrandi S, Scalinci SZ, Mordà D, Rinaldi C, D'Angelo R, Sidoti A. Human retinal secretome: A cross-link between mesenchymal and retinal cells. World J Stem Cells 2023; 15:665-686. [PMID: 37545752 PMCID: PMC10401416 DOI: 10.4252/wjsc.v15.i7.665] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Revised: 03/17/2023] [Accepted: 04/10/2023] [Indexed: 07/25/2023] Open
Abstract
In recent years, mesenchymal stem cells (MSC) have been considered the most effective source for regenerative medicine, especially due to released soluble paracrine bioactive components and extracellular vesicles. These factors, collectively called the secretome, play crucial roles in immunomodulation and in improving survival and regeneration capabilities of injured tissue. Recently, there has been a growing interest in the secretome released by retinal cytotypes, especially retinal pigment epithelium and Müller glia cells. The latter trophic factors represent the key to preserving morphofunctional integrity of the retina, regulating biological pathways involved in survival, function and responding to injury. Furthermore, these factors can play a pivotal role in onset and progression of retinal diseases after damage of cell secretory function. In this review, we delineated the importance of cross-talk between MSCs and retinal cells, focusing on common/induced secreted factors, during experimental therapy for retinal diseases. The cross-link between the MSC and retinal cell secretomes suggests that the MSC secretome can modulate the retinal cell secretome and vice versa. For example, the MSC secretome can protect retinal cells from degeneration by reducing oxidative stress, autophagy and programmed cell death. Conversely, the retinal cell secretome can influence the MSC secretome by inducing changes in MSC gene expression and phenotype.
Collapse
Affiliation(s)
- Luigi Donato
- Department of Biomedical and Dental Sciences and Morphofunctional Imaging, University of Messina, Messina 98125, Italy
- Department of Biomolecular Strategies, Genetics and Cutting-Edge Therapies, Euro-Mediterranean Institute of Science and Technology, Palermo 90139, Italy
| | - Concetta Scimone
- Department of Biomedical and Dental Sciences and Morphofunctional Imaging, University of Messina, Messina 98125, Italy
- Department of Biomolecular Strategies, Genetics and Cutting-Edge Therapies, Euro-Mediterranean Institute of Science and Technology, Palermo 90139, Italy
| | - Simona Alibrandi
- Department of Biomedical and Dental Sciences and Morphofunctional Imaging, University of Messina, Messina 98125, Italy
- Department of Biomolecular Strategies, Genetics and Cutting-Edge Therapies, Euro-Mediterranean Institute of Science and Technology, Palermo 90139, Italy
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Messina 98125, Italy.
| | | | - Domenico Mordà
- Department of Biomedical and Dental Sciences and Morphofunctional Imaging, University of Messina, Messina 98125, Italy
- Department of Biomolecular Strategies, Genetics and Cutting-Edge Therapies, Euro-Mediterranean Institute of Science and Technology, Palermo 90139, Italy
| | - Carmela Rinaldi
- Department of Biomedical and Dental Sciences and Morphofunctional Imaging, University of Messina, Messina 98125, Italy
| | - Rosalia D'Angelo
- Department of Biomedical and Dental Sciences and Morphofunctional Imaging, University of Messina, Messina 98125, Italy
| | - Antonina Sidoti
- Department of Biomedical and Dental Sciences and Morphofunctional Imaging, University of Messina, Messina 98125, Italy
| |
Collapse
|
7
|
Torres-Espin A, Rabadaugh H, Fitzsimons S, Harvey D, Chou A, Lindberg C, Casaletto KB, Goldberger L, Staffaroni AM, Maillard P, Miller BL, DeCarli C, Hinman JD, Ferguson AR, Kramer JH, Elahi FM. Sexually dimorphic differences in angiogenesis markers predict brain aging trajectories. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.07.16.549192. [PMID: 37503183 PMCID: PMC10370093 DOI: 10.1101/2023.07.16.549192] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/29/2023]
Abstract
Aberrant angiogenesis could contribute to cognitive impairment, representing a therapeutic target for preventing dementia. However, most angiogenesis studies focus on model organisms. To test the relevance of angiogenesis to human cognitive aging, we evaluated associations of circulating blood markers of angiogenesis with brain aging trajectories in two deeply phenotyped human cohorts (n=435, age 74 + 9) with longitudinal cognitive assessments, biospecimens, structural brain imaging, and clinical data. Machine learning and traditional statistics revealed sex dimorphic associations of plasma angiogenic growth factors with brain aging outcomes. Specifically, angiogenesis is associated with higher executive function and less brain atrophy in younger women (not men), a directionality of association that reverses around age 75. Higher levels of basic fibroblast growth factor, known for pleiotropic effects on multiple cell types, predicted favorable cognitive trajectories. This work demonstrates the relevance of angiogenesis to brain aging with important therapeutic implications for vascular cognitive impairment and dementia.
Collapse
|
8
|
Huang H, Saddala MS, Mukwaya A, Mohan RR, Lennikov A. Association of Placental Growth Factor and Angiopoietin in Human Retinal Endothelial Cell-Pericyte co-Cultures and iPSC-Derived Vascular Organoids. Curr Eye Res 2023; 48:297-311. [PMID: 36458540 PMCID: PMC10417885 DOI: 10.1080/02713683.2022.2149808] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2022] [Revised: 11/13/2022] [Accepted: 11/14/2022] [Indexed: 12/05/2022]
Abstract
PURPOSE Placental growth factor (PlGF) and Angiopoietin (Ang)-1 are two proteins that are involved in the regulation of endothelial cell (EC) growth and vasculature formation. In the retina and endothelial cells, pericytes are the major source of both molecules. The purpose of this study is to examine the association of PlGF and Ang-1 with human EC/pericyte co-cultures and iPSC-derived vascular organoids. METHODS In this study, we used co-cultures of human primary retinal endothelial cells (HREC) and primary human retinal pericytes (HRP), western blotting, immunofluorescent analysis, TUNEL staining, LDH-assays, and RNA seq analysis, as well as human-induced pluripotent stem cells (iPSC), derived organoids (VO) to study the association between PlGF and Ang-1. RESULTS Inhibition of PlGF by PlGF neutralizing antibody in HREC-HRP co-cultures resulted in the increased expression of Ang-1 and Tie-2 in a dose-dependent manner. This upregulation was not observed in HREC and HRP monocultures but only in co-cultures suggesting the association of pericytes and endothelial cells. Furthermore, Vascular endothelial growth factor receptor 1 (VEGFR1) inhibition abolished the Ang-1 and Tie-2 upregulation by PlGF inhibition. The pericyte viability in high-glucose conditions was also reduced by VEGFR1 neutralization. Immunofluorescent analysis showed that Ang-1 and Ang-2 were expressed mainly by perivascular cells in the VO. RNA seq analysis of the RNA isolated from VO in high glucose conditions indicated increased PlGF and Ang-2 expressions in the VO. PlGF inhibition increased the expression of Ang-1 and Tie-2 in VO, increasing the pericyte coverage of the VO microvascular network. CONCLUSION Combined, these results suggest PlGF's role in the regulation of Ang-1 and Tie-2 expression through VEGFR1. These findings provide new insights into the neovascularization process in diabetic retinopathy and new targets for potential therapeutic intervention.
Collapse
Affiliation(s)
- Hu Huang
- Department of Ophthalmology, University of Missouri School of Medicine, Columbia, MO, USA
| | - Madhu Sudhana Saddala
- Department of Ophthalmology, University of Missouri School of Medicine, Columbia, MO, USA
- Wilmer Bioinformatics, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Anthony Mukwaya
- Department of Ophthalmology, Institute for Clinical, and Experimental Medicine, Faculty of Health Sciences, Linköping University, Linköping, Sweden
- Department of Biochemistry and Molecular Biology, Faculty of Health Sciences, Busitema University, Uganda
| | - Rajiv R. Mohan
- Department of Ophthalmology, University of Missouri School of Medicine, Columbia, MO, USA
| | - Anton Lennikov
- Department of Ophthalmology, Schepens Eye Research Institute of Massachusetts Eye and Ear, Harvard Medical School, Boston, Massachusetts, United States
| |
Collapse
|
9
|
Hwang JS, Shin YJ. Role of Choline in Ocular Diseases. Int J Mol Sci 2021; 22:4733. [PMID: 33946979 PMCID: PMC8124599 DOI: 10.3390/ijms22094733] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Revised: 04/25/2021] [Accepted: 04/28/2021] [Indexed: 12/29/2022] Open
Abstract
Choline is essential for maintaining the structure and function of cells in humans. Choline plays an important role in eye health and disease. It is a precursor of acetylcholine, a neurotransmitter of the parasympathetic nervous system, and it is involved in the production and secretion of tears by the lacrimal glands. It also contributes to the stability of the cells and tears on the ocular surface and is involved in retinal development and differentiation. Choline deficiency is associated with retinal hemorrhage, glaucoma, and dry eye syndrome. Choline supplementation may be effective for treating these diseases.
Collapse
Affiliation(s)
| | - Young-Joo Shin
- Department of Ophthalmology, Hallym University Medical Center, Hallym University College of Medicine, Seoul 07442, Korea;
| |
Collapse
|