1
|
Huang C, Zhang X, Wu M, Yang C, Ge X, Chen W, Li X, Liu S, Yang S. IL-1β-induced pericyte dysfunction with a secretory phenotype exacerbates retinal microenvironment inflammation via Hes1/STAT3 signaling pathway. Int Immunopharmacol 2025; 144:113611. [PMID: 39612772 DOI: 10.1016/j.intimp.2024.113611] [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: 07/22/2024] [Revised: 11/06/2024] [Accepted: 11/07/2024] [Indexed: 12/01/2024]
Abstract
Retinal pericytes are mural cells surrounding capillaries to maintain the integrity of blood-retina barrier and regulate vascular behaviors. Pericyte loss has been considered as the hallmark of diabetic retinopathy (DR), which is a major complication of diabetes and the leading cause of blindness in adults. However, the precise function of pericytes in regulating the retinal microenvironment and the underlying mechanism remains largely unknown. In this study, we observed a secretory phenotype of pericytes with elevated inflammatory cytokines in response to Interleukin-1β (IL-1β), a canonical inflammatory cytokine which significantly increases during the initial stages of diabetic retinopathy. This phenotype is also accompanied by reduced expression of adherent junction proteins and contractile proteins. Paracrine cytokines derived from pericytes further induce the chemotaxis of microglia cells and trigger detrimental changes in endothelial cells, including reduced expression of tight junction protein Occludin and increased apoptosis. Mechanically, the secretion potential in pericytes is partially mediated by Hes1/STAT3 signaling pathway. Moreover, co-injection of stattic, an inhibitor targeting STAT3 activation, could effectively attenuate IL-1β-induced retinal inflammation and microglial activation in retina tissues. Collectively, these findings demonstrate the potential of retinal pericytes as an initial inflammatory sensor prior to their anatomical pathological loss, via undergoing phenotypic changes and secreting paracrine factors to amplify local inflammation and damage endothelial cells in vitro. Furthermore, inhibition of STAT3 activation by inhibitors significantly ameliorates IL-1β-induced retinal inflammation, suggesting STAT3 in retinal pericytes as a promising target for alleviating DR and other IL-1β-induced ocular diseases.
Collapse
Affiliation(s)
- Caoxin Huang
- Xiamen Diabetes Institute, Fujian Province Key Laboratory of Translational Research for Diabetes, Department of Endocrinology and Diabetes, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, China.
| | - Xiaofang Zhang
- Xiamen Diabetes Institute, Fujian Province Key Laboratory of Translational Research for Diabetes, Department of Endocrinology and Diabetes, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, China
| | - Menghua Wu
- Xiamen Diabetes Institute, Fujian Province Key Laboratory of Translational Research for Diabetes, Department of Endocrinology and Diabetes, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, China
| | - Chen Yang
- Xiamen Diabetes Institute, Fujian Province Key Laboratory of Translational Research for Diabetes, Department of Endocrinology and Diabetes, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, China
| | - Xilin Ge
- Xiamen Diabetes Institute, Fujian Province Key Laboratory of Translational Research for Diabetes, Department of Endocrinology and Diabetes, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, China
| | - Wenting Chen
- Xiamen Diabetes Institute, Fujian Province Key Laboratory of Translational Research for Diabetes, Department of Endocrinology and Diabetes, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, China
| | - Xuejun Li
- Xiamen Diabetes Institute, Fujian Province Key Laboratory of Translational Research for Diabetes, Department of Endocrinology and Diabetes, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, China
| | - Suhuan Liu
- Xiamen Diabetes Institute, Fujian Province Key Laboratory of Translational Research for Diabetes, Department of Endocrinology and Diabetes, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, China; Research Center for Translational Medicine, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, China.
| | - Shuyu Yang
- Xiamen Diabetes Institute, Fujian Province Key Laboratory of Translational Research for Diabetes, Department of Endocrinology and Diabetes, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, China; Research Studio of Traditional Chinese Medicine, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, China.
| |
Collapse
|
2
|
Gregg AT, Wang T, Szczepan M, Lam E, Yagi H, Neilsen K, Wang X, Smith LEH, Sun Y. Botulinum neurotoxin serotype A inhibited ocular angiogenesis through modulating glial activation via SOCS3. Angiogenesis 2024; 27:753-764. [PMID: 38922557 PMCID: PMC11564384 DOI: 10.1007/s10456-024-09935-7] [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: 03/28/2024] [Accepted: 06/18/2024] [Indexed: 06/27/2024]
Abstract
BACKGROUND Pathological angiogenesis causes significant vision loss in neovascular age-related macular degeneration and other retinopathies with neovascularization (NV). Neuronal/glial-vascular interactions influence the release of angiogenic and neurotrophic factors. We hypothesized that botulinum neurotoxin serotype A (BoNT/A) modulates pathological endothelial cell proliferation through glial cell activation and growth factor release. METHODS A laser-induced choroidal NV (CNV) was employed to investigate the anti-angiogenic effects of BoNT/A. Fundus fluorescence angiography, immunohistochemistry, and real-time PCR were used to assess BoNT/A efficacy in inhibiting CNV and the molecular mechanisms underlying this inhibition. Neuronal and glial suppressor of cytokine signaling 3 (SOCS3) deficient mice were used to investigate the molecular mechanisms of BoNT/A in inhibiting CNV via SOCS3. FINDINGS In laser-induced CNV mice with intravitreal BoNT/A treatment, CNV lesions decreased > 30%; vascular leakage and retinal glial activation were suppressed; and Socs3 mRNA expression was induced while vascular endothelial growth factor A (Vegfa) mRNA expression was suppressed. The protective effects of BoNT/A on CNV development were diminished in mice lacking neuronal/glial SOCS3. CONCLUSION BoNT/A suppressed laser-induced CNV and glial cell activation, in part through SOCS3 induction in neuronal/glial cells. BoNT/A treatment led to a decrease of pro-angiogenic factors, including VEGFA, highlighting the potential of BoNT/A as a therapeutic intervention for pathological angiogenesis in retinopathies.
Collapse
Affiliation(s)
- Austin T Gregg
- Department of Ophthalmology, Boston Children's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Tianxi Wang
- Department of Ophthalmology, Boston Children's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Manon Szczepan
- Department of Ophthalmology, Boston Children's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Enton Lam
- Department of Ophthalmology, Boston Children's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Hitomi Yagi
- Department of Ophthalmology, Boston Children's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Katherine Neilsen
- Department of Ophthalmology, Boston Children's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Xingyan Wang
- Department of Ophthalmology, Boston Children's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Lois E H Smith
- Department of Ophthalmology, Boston Children's Hospital, Harvard Medical School, Boston, MA, 02115, USA.
| | - Ye Sun
- Department of Ophthalmology, Boston Children's Hospital, Harvard Medical School, Boston, MA, 02115, USA.
| |
Collapse
|
3
|
Wang X, Wang T, Kaneko S, Kriukov E, Lam E, Szczepan M, Chen J, Gregg A, Wang X, Fernandez-Gonzalez A, Mitsialis SA, Kourembanas S, Baranov P, Sun Y. Photoreceptors inhibit pathological retinal angiogenesis through transcriptional regulation of Adam17 via c-Fos. Angiogenesis 2024; 27:379-395. [PMID: 38483712 PMCID: PMC11303108 DOI: 10.1007/s10456-024-09912-0] [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: 11/14/2023] [Accepted: 03/03/2024] [Indexed: 04/11/2024]
Abstract
Pathological retinal angiogenesis profoundly impacts visual function in vascular eye diseases, such as retinopathy of prematurity (ROP) in preterm infants and age-related macular degeneration in the elderly. While the involvement of photoreceptors in these diseases is recognized, the underlying mechanisms remain unclear. This study delved into the pivotal role of photoreceptors in regulating abnormal retinal blood vessel growth using an oxygen-induced retinopathy (OIR) mouse model through the c-Fos/A disintegrin and metalloprotease 17 (Adam17) axis. Our findings revealed a significant induction of c-Fos expression in rod photoreceptors, and c-Fos depletion in these cells inhibited pathological neovascularization and reduced blood vessel leakage in the OIR mouse model. Mechanistically, c-Fos directly regulated the transcription of Adam17 a shedding protease responsible for the production of bioactive molecules involved in inflammation, angiogenesis, and cell adhesion and migration. Furthermore, we demonstrated the therapeutic potential by using an adeno-associated virus carrying a rod photoreceptor-specific short hairpin RNA against c-fos which effectively mitigated abnormal retinal blood vessel overgrowth, restored retinal thickness, and improved electroretinographic (ERG) responses. In conclusion, this study highlights the significance of photoreceptor c-Fos in ROP pathology, offering a novel perspective for the treatment of this disease.
Collapse
Affiliation(s)
- Xudong Wang
- Department of Ophthalmology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Tianxi Wang
- Department of Ophthalmology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Satoshi Kaneko
- Department of Ophthalmology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Emil Kriukov
- Department of Ophthalmology, The Schepens Eye Research Institute of Massachusetts Eye and Ear, Harvard Medical School, Boston, MA, USA
| | - Enton Lam
- Department of Ophthalmology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Manon Szczepan
- Department of Ophthalmology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Jasmine Chen
- Department of Ophthalmology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Austin Gregg
- Department of Ophthalmology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Xingyan Wang
- Department of Ophthalmology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Angeles Fernandez-Gonzalez
- Division of Newborn Medicine, Department of Pediatrics, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - S Alex Mitsialis
- Division of Newborn Medicine, Department of Pediatrics, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Stella Kourembanas
- Division of Newborn Medicine, Department of Pediatrics, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Petr Baranov
- Department of Ophthalmology, The Schepens Eye Research Institute of Massachusetts Eye and Ear, Harvard Medical School, Boston, MA, USA
| | - Ye Sun
- Department of Ophthalmology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
4
|
Ahmed CM, Johnson HM, Lewin AS. Corneal application of SOCS1/3 peptides for the treatment of eye diseases mediated by inflammation and oxidative stress. Front Immunol 2024; 15:1416181. [PMID: 39104531 PMCID: PMC11298391 DOI: 10.3389/fimmu.2024.1416181] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Accepted: 07/05/2024] [Indexed: 08/07/2024] Open
Abstract
Several blinding diseases affecting the retina and optic nerve are exacerbated by or caused by dysregulated inflammation and oxidative stress. These diseases include uveitis, age related macular degeneration, diabetic retinopathy and glaucoma. Consequently, despite their divergent symptoms, treatments that reduce oxidative stress and suppress inflammation may be therapeutic. The production of inflammatory cytokines and their activities are regulated by a class of proteins termed Suppressors of Cytokine Signaling (SOCS). SOCS1 and SOCS3 are known to dampen signaling via pathways employing Janus kinases and signal transducer and activator of transcription proteins (JAK/STAT), Toll-like Receptors (TLR), nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB), mitogen activated kinase (MAPK) and NLR family pyrin domain containing 3 (NLRP3). We have developed cell-penetrating peptides from the kinase inhibitory region of the SOCS1 and SOCS3 (denoted as R9-SOCS1-KIR and R9-SOCS3-KIR) and tested them in retinal pigment epithelium (RPE) cells and in macrophage cell lines. SOCS-KIR peptides exhibited anti-inflammatory, anti-oxidant and anti-angiogenic properties. In cell culture, both Th1 and Th17 cells were suppressed together with the inhibition of other inflammatory markers. We also observed a decrease in oxidants and a simultaneous rise in neuroprotective and anti-oxidant effectors. In addition, treatment prevented the loss of gap junction proteins and the ensuing drop in transepithelial electrical resistance in RPE cells. When tested in mouse models by eye drop instillation, they showed protection against autoimmune uveitis, as a prophylactic as well as a therapeutic. Mice with endotoxin-induced uveitis were protected by eye drop administration as well. R9-SOCS3-KIR was particularly effective against the pathways acting through STAT3, e.g. IL-6 and VEGF-A mediated responses that lead to macular degeneration. Eye drop administration of R9-SOCS3-KIR stimulated production of antioxidant effectors and reduced clinical symptoms in mouse model of oxidative stress that replicates the RPE injury occurring in AMD. Because these peptides suppress multiple pathogenic stimuli and because they can be delivered topically to the cornea, they are attractive candidates for therapeutics for uveitis, macular degeneration, diabetic retinopathy and glaucoma.
Collapse
Affiliation(s)
- Chulbul M. Ahmed
- Department of Molecular Genetics and Microbiology, University of Florida, Gainesville, FL, United States
| | - Howard M. Johnson
- Department of Microbiology and Cell Science, University of Florida, Gainesville, FL, United States
| | - Alfred S. Lewin
- Department of Molecular Genetics and Microbiology, University of Florida, Gainesville, FL, United States
| |
Collapse
|
5
|
Wang T, Kaneko S, Kriukov E, Alvarez D, Lam E, Wang Y, La Manna S, Marasco D, Fernandez-Gonzalez A, Mitsialis SA, Kourembanas S, Stahl A, Chen M, Xu H, Baranov P, Cai G, von Andrian UH, Sun Y. SOCS3 regulates pathological retinal angiogenesis through modulating SPP1 expression in microglia and macrophages. Mol Ther 2024; 32:1425-1444. [PMID: 38504518 PMCID: PMC11081920 DOI: 10.1016/j.ymthe.2024.03.025] [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: 11/01/2023] [Revised: 02/18/2024] [Accepted: 03/15/2024] [Indexed: 03/21/2024] Open
Abstract
Pathological ocular angiogenesis has long been associated with myeloid cell activation. However, the precise cellular and molecular mechanisms governing the intricate crosstalk between the immune system and vascular changes during ocular neovascularization formation remain elusive. In this study, we demonstrated that the absence of the suppressor of cytokine signaling 3 (SOCS3) in myeloid cells led to a substantial accumulation of microglia and macrophage subsets during the neovascularization process. Our single-cell RNA sequencing data analysis revealed a remarkable increase in the expression of the secreted phosphoprotein 1 (Spp1) gene within these microglia and macrophages, identifying subsets of Spp1-expressing microglia and macrophages during neovascularization formation in angiogenesis mouse models. Notably, the number of Spp1-expressing microglia and macrophages exhibited further elevation during neovascularization in mice lacking myeloid SOCS3. Moreover, our investigation unveiled the Spp1 gene as a direct transcriptional target gene of signal transducer and activator of transcription 3. Importantly, pharmaceutical activation of SOCS3 or blocking of SPP1 resulted in a significant reduction in pathological neovascularization. In conclusion, our study highlights the pivotal role of the SOCS3/STAT3/SPP1 axis in the regulation of pathological retinal angiogenesis.
Collapse
Affiliation(s)
- Tianxi Wang
- Department of Ophthalmology, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Satoshi Kaneko
- Department of Ophthalmology, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Emil Kriukov
- Department of Ophthalmology, The Schepens Eye Research Institute of Massachusetts Eye and Ear, Harvard Medical School, Boston, MA 02114, USA
| | - David Alvarez
- Department of Immunology and HMS Center for Immune Imaging, Harvard Medical School, Boston, MA 02115, USA
| | - Enton Lam
- Department of Ophthalmology, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Yidi Wang
- Department of Immunology and HMS Center for Immune Imaging, Harvard Medical School, Boston, MA 02115, USA
| | - Sara La Manna
- Department of Pharmacy, University of Naples "Federico II", 80138 Naples, Italy
| | - Daniela Marasco
- Department of Pharmacy, University of Naples "Federico II", 80138 Naples, Italy
| | - Angeles Fernandez-Gonzalez
- Division of Newborn Medicine, Department of Pediatrics, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - S Alex Mitsialis
- Division of Newborn Medicine, Department of Pediatrics, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Stella Kourembanas
- Division of Newborn Medicine, Department of Pediatrics, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Andreas Stahl
- Department of Ophthalmology, University Medicine Greifswald, 17475 Greifswald, Germany
| | - Mei Chen
- Centre for Experimental Medicine, School of Medicine, Dentistry and Biomedical Sciences, Queen's University Belfast, Belfast, UK
| | - Heping Xu
- Centre for Experimental Medicine, School of Medicine, Dentistry and Biomedical Sciences, Queen's University Belfast, Belfast, UK
| | - Petr Baranov
- Department of Ophthalmology, The Schepens Eye Research Institute of Massachusetts Eye and Ear, Harvard Medical School, Boston, MA 02114, USA
| | - Guoshuai Cai
- Department of Surgery, College of Medicine, University of Florida, Gainesville, FL 32610, USA
| | - Ulrich H von Andrian
- Department of Immunology and HMS Center for Immune Imaging, Harvard Medical School, Boston, MA 02115, USA; The Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA 02139, USA
| | - Ye Sun
- Department of Ophthalmology, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
6
|
Tzani A, Haemmig S, Cheng HS, Perez-Cremades D, Augusto Heuschkel M, Jamaiyar A, Singh S, Aikawa M, Yu P, Wang T, Ye S, Feinberg MW, Plutzky J. FAM222A, Part of the BET-Regulated Basal Endothelial Transcriptome, Is a Novel Determinant of Endothelial Biology and Angiogenesis-Brief Report. Arterioscler Thromb Vasc Biol 2024; 44:143-155. [PMID: 37942611 PMCID: PMC10840377 DOI: 10.1161/atvbaha.123.319909] [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: 07/22/2023] [Accepted: 10/17/2023] [Indexed: 11/10/2023]
Abstract
BACKGROUND BETs (bromodomain and extraterminal domain-containing epigenetic reader proteins), including BRD4 (bromodomain-containing protein 4), orchestrate transcriptional programs induced by pathogenic stimuli, as intensively studied in cardiovascular disease and elsewhere. In endothelial cells (ECs), BRD4 directs induced proinflammatory, proatherosclerotic transcriptional responses; BET inhibitors, like JQ1, repress these effects and decrease atherosclerosis. While BET effects in pathogenic conditions have prompted therapeutic BET inhibitor development, BET action under basal conditions, including ECs, has remained understudied. To understand BET action in basal endothelial transcriptional programs, we first analyzed EC RNA-Seq data in the absence versus presence of JQ1 before using BET regulation to identify novel determinants of EC biology and function. METHODS RNA-Seq datasets of human umbilical vein ECs without and with JQ1 treatment were analyzed. After identifying C12orf34, also known as FAM222A (family with sequence similarity 222 member A), as a previously unreported, basally expressed, potently JQ1-induced EC gene, FAM222A was studied in endothelial and angiogenic responses in vitro using small-interference RNA silencing and lentiviral overexpression, in vitro, ex vivo and in vivo, including aortic sprouting, matrigel plug assays, and murine neonatal oxygen-induced retinopathy. RESULTS Resting EC RNA-Seq data indicate BETs direct transcriptional programs underlying core endothelial properties including migration, proliferation, and angiogenesis. BET inhibition in resting ECs also significantly induced a subset of mRNAs, including FAM222A-a unique BRD4-regulated gene with no reported EC role. Silencing endothelial FAM222A significantly decreased cellular proliferation, migration, network formation, aorta sprouting, and Matrigel plug vascularization through coordinated modulation of VEGF (vascular endothelial growth factor) and NOTCH mediator expression in vitro, ex vivo, in vivo; lentiviral FAM222A overexpression had opposite effects. In vivo, siFAM222A significantly repressed retinal revascularization in neonatal murine oxygen-induced retinopathy through similar angiogenic signaling modulation. CONCLUSIONS BET control over the basal endothelial transcriptome includes FAM222A, a novel, BRD4-regulated, key determinant of endothelial biology and angiogenesis.
Collapse
Affiliation(s)
- Aspasia Tzani
- Division of Cardiovascular Medicine, Brigham and Women’s Hospital, Boston, MA 02115
- Harvard Medical School, Boston, MA 02115
| | - Stefan Haemmig
- Division of Cardiovascular Medicine, Brigham and Women’s Hospital, Boston, MA 02115
- Harvard Medical School, Boston, MA 02115
| | - Henry S. Cheng
- Division of Cardiovascular Medicine, Brigham and Women’s Hospital, Boston, MA 02115
- Harvard Medical School, Boston, MA 02115
| | - Daniel Perez-Cremades
- Division of Cardiovascular Medicine, Brigham and Women’s Hospital, Boston, MA 02115
- Harvard Medical School, Boston, MA 02115
| | - Marina Augusto Heuschkel
- Division of Cardiovascular Medicine, Brigham and Women’s Hospital, Boston, MA 02115
- Harvard Medical School, Boston, MA 02115
| | - Anurag Jamaiyar
- Division of Cardiovascular Medicine, Brigham and Women’s Hospital, Boston, MA 02115
- Harvard Medical School, Boston, MA 02115
| | - Sasha Singh
- Division of Cardiovascular Medicine, Brigham and Women’s Hospital, Boston, MA 02115
- Harvard Medical School, Boston, MA 02115
| | - Masanori Aikawa
- Division of Cardiovascular Medicine, Brigham and Women’s Hospital, Boston, MA 02115
- Harvard Medical School, Boston, MA 02115
| | - Paul Yu
- Division of Cardiovascular Medicine, Brigham and Women’s Hospital, Boston, MA 02115
- Harvard Medical School, Boston, MA 02115
| | - Tianxi Wang
- Department of Ophthalmology, Boston Children’s Hospital, Boston, MA 02115
- Harvard Medical School, Boston, MA 02115
| | - Sun Ye
- Department of Ophthalmology, Boston Children’s Hospital, Boston, MA 02115
- Harvard Medical School, Boston, MA 02115
| | - Mark W. Feinberg
- Division of Cardiovascular Medicine, Brigham and Women’s Hospital, Boston, MA 02115
- Harvard Medical School, Boston, MA 02115
| | - Jorge Plutzky
- Division of Cardiovascular Medicine, Brigham and Women’s Hospital, Boston, MA 02115
- Harvard Medical School, Boston, MA 02115
| |
Collapse
|
7
|
Ahmed CM, Patel AP, Johnson HM, Ildefonso CJ, Lewin AS. Suppressor of cytokine signaling 3-derived peptide as a therapeutic for inflammatory and oxidative stress-induced damage to the retina. Mol Vis 2023; 29:338-356. [PMID: 38264613 PMCID: PMC10805335] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Accepted: 12/15/2023] [Indexed: 01/25/2024] Open
Abstract
Purpose Inflammation and oxidative stress contribute to age-related macular degeneration (AMD) and other retinal diseases. We tested a cell-penetrating peptide from the kinase inhibitory region of an intracellular checkpoint inhibitor suppressor of cytokine signaling 3 (R9-SOCS3-KIR) peptide for its ability to blunt the inflammatory or oxidative pathways leading to AMD. Methods We used anaphylatoxin C5a to mimic the effect of activated complement, lipopolysaccharide (LPS), and tumor necrosis factor alpha (TNFα) to stimulate inflammation and paraquat to induce mitochondrial oxidative stress. We used a human retinal pigment epithelium (RPE) cell line (ARPE-19) as proliferating cells and a mouse macrophage cell line (J774A.1) to follow cell propagation using microscopy or cell titer assays. We evaluated inflammatory pathways by monitoring the nuclear translocation of NF-κB p65 and mitogen-activated protein kinase p38. Quantitative reverse transcription polymerase chain reaction (qRT-PCR) and Western blot were used to evaluate the induction of inflammatory markers. In differentiated ARPE-19 monolayers, we evaluated the integrity of tight junction proteins through microscopy and the measurement of transepithelial electrical resistance (TEER). We used intraperitoneal injection of sodium iodate in mice to test the ability of R9-SOC3-KIR to prevent RPE and retinal injury, as assessed by fundoscopy, optical coherence tomography, and histology. Results R9-SOCS3-KIR treatment suppressed C5a-induced nuclear translocation of the NF-kB activation domain p65 in undifferentiated ARPE-19 cells. TNF-mediated damage to tight junction proteins in RPE, and the loss of TEER was prevented in the presence of R9-SOCS3-KIR. Treatment with the R9-SOCS3-KIR peptide blocked the C5a-induced expression of inflammatory genes. The R9-SOCS3-KIR treatment also blocked the LPS-induced expression of interleukin-6, MCP1, cyclooxygenase 2, and interleukin-1 beta. R9-SOCS3-KIR prevented paraquat-mediated cell death and enhanced the levels of antioxidant effectors. Daily eye drop treatment with R9-SOCS3-KIR protected against retinal injury caused by i.p. administration of sodium iodate. Conclusions R9-SOCS3-KIR blocks the induction of inflammatory signaling in cell culture and reduces retinal damage in a widely used RPE/retinal oxidative injury model. As this peptide can be administered through corneal instillation, this treatment may offer a convenient way to slow down the progression of ocular diseases arising from inflammation and chronic oxidative stress.
Collapse
Affiliation(s)
- Chulbul M Ahmed
- Department of Molecular Genetics and Microbiology, University of Florida Gainesville, FL
| | - Anil P Patel
- Department of Molecular Genetics and Microbiology, University of Florida Gainesville, FL
| | - Howard M Johnson
- Department of Microbiology and Cell Science, University of Florida Gainesville, FL
| | | | - Alfred S Lewin
- Department of Molecular Genetics and Microbiology, University of Florida Gainesville, FL
| |
Collapse
|
8
|
Yoshimoto T, Chaya T, Varner LR, Ando M, Tsujii T, Motooka D, Kimura K, Furukawa T. The Rax homeoprotein in Müller glial cells is required for homeostasis maintenance of the postnatal mouse retina. J Biol Chem 2023; 299:105461. [PMID: 37977220 PMCID: PMC10714373 DOI: 10.1016/j.jbc.2023.105461] [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: 07/20/2023] [Revised: 10/25/2023] [Accepted: 11/04/2023] [Indexed: 11/19/2023] Open
Abstract
Müller glial cells, which are the most predominant glial subtype in the retina, play multiple important roles, including the maintenance of structural integrity, homeostasis, and physiological functions of the retina. We have previously found that the Rax homeoprotein is expressed in postnatal and mature Müller glial cells in the mouse retina. However, the function of Rax in postnatal and mature Müller glial cells remains to be elucidated. In the current study, we first investigated Rax function in retinal development using retroviral lineage analysis and found that Rax controls the specification of late-born retinal cell types, including Müller glial cells in the postnatal retina. We next generated Rax tamoxifen-induced conditional KO (Rax iCKO) mice, where Rax can be depleted in mTFP-labeled Müller glial cells upon tamoxifen treatment, by crossing Raxflox/flox mice with Rlbp1-CreERT2 mice, which we have produced. Immunohistochemical analysis showed a characteristic of reactive gliosis and enhanced gliosis of Müller glial cells in Rax iCKO retinas under normal and stress conditions, respectively. We performed RNA-seq analysis on mTFP-positive cells purified from the Rax iCKO retina and found significantly reduced expression of suppressor of cytokinesignaling-3 (Socs3). Reporter gene assays showed that Rax directly transactivates the Socs3 promoter. We observed decreased expression of Socs3 in Müller glial cells of Rax iCKO retinas by immunostaining. Taken together, the present results suggest that Rax suppresses inflammation in Müller glial cells by transactivating Socs3. This study sheds light on the transcriptional regulatory mechanisms underlying retinal Müller glial cell homeostasis.
Collapse
Affiliation(s)
- Takuya Yoshimoto
- Laboratory for Molecular and Developmental Biology, Institute for Protein Research, Osaka University, Suita, Osaka, Japan; Department of Ophthalmology, Yamaguchi University Graduate School of Medicine, Yamaguchi University, Ube, Yamaguchi, Japan
| | - Taro Chaya
- Laboratory for Molecular and Developmental Biology, Institute for Protein Research, Osaka University, Suita, Osaka, Japan
| | - Leah R Varner
- Laboratory for Molecular and Developmental Biology, Institute for Protein Research, Osaka University, Suita, Osaka, Japan
| | - Makoto Ando
- Laboratory for Molecular and Developmental Biology, Institute for Protein Research, Osaka University, Suita, Osaka, Japan
| | - Toshinori Tsujii
- Laboratory for Molecular and Developmental Biology, Institute for Protein Research, Osaka University, Suita, Osaka, Japan
| | - Daisuke Motooka
- Genome Information Research Center, Research Institute for Microbial Diseases, Osaka University, Osaka, Japan
| | - Kazuhiro Kimura
- Department of Ophthalmology, Yamaguchi University Graduate School of Medicine, Yamaguchi University, Ube, Yamaguchi, Japan
| | - Takahisa Furukawa
- Laboratory for Molecular and Developmental Biology, Institute for Protein Research, Osaka University, Suita, Osaka, Japan.
| |
Collapse
|
9
|
Cantu A, Gutierrez MC, Dong X, Leek C, Anguera M, Lingappan K. Modulation of recovery from neonatal hyperoxic lung injury by sex as a biological variable. Redox Biol 2023; 68:102933. [PMID: 38661305 PMCID: PMC10628633 DOI: 10.1016/j.redox.2023.102933] [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: 08/09/2023] [Revised: 10/07/2023] [Accepted: 10/11/2023] [Indexed: 04/26/2024] Open
Abstract
Recovery from lung injury during the neonatal period requires the orchestration of many biological pathways. The modulation of such pathways can drive the developing lung towards proper repair or persistent maldevelopment that can lead to a disease phenotype. Sex as a biological variable can regulate these pathways differently in the male and female lung exposed to neonatal hyperoxia. In this study, we assessed the contribution of cellular diversity in the male and female neonatal lung following injury. Our objective was to investigate sex and cell-type specific transcriptional changes that drive repair or persistent injury in the neonatal lung and delineate the alterations in the immune-endothelial cell communication networks using single cell RNA sequencing (sc-RNAseq) in a murine model of hyperoxic injury. We generated transcriptional profiles of >55,000 cells isolated from the lungs of postnatal day 1 (PND 1; pre-exposure), PND 7, and PND 21neonatal male and female C57BL/6 mice exposed to 95 % FiO2 between PND 1-5 (saccular stage of lung development). We show the presence of sex-based differences in the transcriptional states of lung endothelial and immune cells at PND 1 and PND 21. Furthermore, we demonstrate that biological sex significantly influences the response to injury, with a greater number of differentially expressed genes showing sex-specific patterns than those shared between male and female lungs. Pseudotime trajectory analysis highlighted genes needed for lung development that were altered by hyperoxia. Finally, we show intercellular communication between endothelial and immune cells at saccular and alveolar stages of lung development with sex-based biases in the crosstalk and identify novel ligand-receptor pairs. Our findings provide valuable insights into the cell diversity, transcriptional state, developmental trajectory, and cell-cell communication underlying neonatal lung injury, with implications for understanding lung development and possible therapeutic interventions while highlighting the crucial role of sex as a biological variable.
Collapse
Affiliation(s)
- Abiud Cantu
- Department of Neonatology, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | | | - Xiaoyu Dong
- Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA
| | - Connor Leek
- Department of Neonatology, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Montserrat Anguera
- Department of Biomedical Sciences, University of Pennsylvania School of Veterinary Medicine, Philadelphia, PA, USA
| | - Krithika Lingappan
- Department of Neonatology, Children's Hospital of Philadelphia, Philadelphia, PA, USA.
| |
Collapse
|
10
|
Cantu A, Gutierrez MC, Dong X, Leek C, Anguera M, Lingappan K. Modulation of Recovery from Neonatal Hyperoxic Lung Injury by Sex as a Biological Variable. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.08.09.552532. [PMID: 37609288 PMCID: PMC10441379 DOI: 10.1101/2023.08.09.552532] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/24/2023]
Abstract
Recovery from lung injury during the neonatal period requires the orchestration of many biological pathways. The modulation of such pathways can drive the developing lung towards proper repair or persistent maldevelopment that can lead to a disease phenotype. Sex as a biological variable can regulate these pathways differently in the male and female lung exposed to neonatal hyperoxia. In this study, we assessed the contribution of cellular diversity in the male and female neonatal lung following injury. Our objective was to investigate sex and cell-type specific transcriptional changes that drive repair or persistent injury in the neonatal lung and delineate the alterations in the immune-endothelial cell communication networks using single cell RNA sequencing (sc-RNAseq) in a murine model of hyperoxic injury. We generated transcriptional profiles of >55,000 cells isolated from the lungs of postnatal day 1 (PND 1) and postnatal day 21 (PND 21) neonatal male and female C57BL/6 mice exposed to 95% FiO 2 between PND 1-5 (saccular stage of lung development). We show the presence of sex-based differences in the transcriptional states of lung endothelial and immune cells at PND 1 and PND 21. Furthermore, we demonstrate that biological sex significantly influences the response to injury, with a greater number of differentially expressed genes showing sex-specific patterns than those shared between male and female lungs. Pseudotime trajectory analysis highlighted genes needed for lung development that were altered by hyperoxia. Finally, we show intercellular communication between endothelial and immune cells at saccular and alveolar stages of lung development with sex-based biases in the crosstalk and identify novel ligand-receptor pairs. Our findings provide valuable insights into the cell diversity, transcriptional state, developmental trajectory, and cell-cell communication underlying neonatal lung injury, with implications for understanding lung development and possible therapeutic interventions while highlighting the crucial role of sex as a biological variable.
Collapse
|
11
|
Wang X, Wang T, Lam E, Alvarez D, Sun Y. Ocular Vascular Diseases: From Retinal Immune Privilege to Inflammation. Int J Mol Sci 2023; 24:12090. [PMID: 37569464 PMCID: PMC10418793 DOI: 10.3390/ijms241512090] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Revised: 07/21/2023] [Accepted: 07/26/2023] [Indexed: 08/13/2023] Open
Abstract
The eye is an immune privileged tissue that insulates the visual system from local and systemic immune provocation to preserve homeostatic functions of highly specialized retinal neural cells. If immune privilege is breached, immune stimuli will invade the eye and subsequently trigger acute inflammatory responses. Local resident microglia become active and release numerous immunological factors to protect the integrity of retinal neural cells. Although acute inflammatory responses are necessary to control and eradicate insults to the eye, chronic inflammation can cause retinal tissue damage and cell dysfunction, leading to ocular disease and vision loss. In this review, we summarized features of immune privilege in the retina and the key inflammatory responses, factors, and intracellular pathways activated when retinal immune privilege fails, as well as a highlight of the recent clinical and research advances in ocular immunity and ocular vascular diseases including retinopathy of prematurity, age-related macular degeneration, and diabetic retinopathy.
Collapse
Affiliation(s)
- Xudong Wang
- Department of Ophthalmology, Harvard Medical School, Boston Children’s Hospital, Boston, MA 02115, USA; (X.W.)
| | - Tianxi Wang
- Department of Ophthalmology, Harvard Medical School, Boston Children’s Hospital, Boston, MA 02115, USA; (X.W.)
| | - Enton Lam
- Department of Ophthalmology, Harvard Medical School, Boston Children’s Hospital, Boston, MA 02115, USA; (X.W.)
| | - David Alvarez
- Department of Immunology, Harvard Medical School, Boston, MA 02115, USA
| | - Ye Sun
- Department of Ophthalmology, Harvard Medical School, Boston Children’s Hospital, Boston, MA 02115, USA; (X.W.)
| |
Collapse
|
12
|
Hu J, Lin F, Yin Y, Shang Y, Xiao Z, Xu W. Adipocyte-derived exosomal miR-30c-5p promotes ovarian angiogenesis in polycystic ovary syndrome via the SOCS3/STAT3/VEGFA pathway. J Steroid Biochem Mol Biol 2023; 230:106278. [PMID: 36870372 DOI: 10.1016/j.jsbmb.2023.106278] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/02/2023] [Revised: 02/07/2023] [Accepted: 03/02/2023] [Indexed: 03/06/2023]
Abstract
Polycystic ovary syndrome (PCOS) is a systemic endocrine disease affecting women's reproductive health. Ovarian angiogenesis in PCOS patients is abnormal, manifested by increased ovarian stromal vascularization and upregulated proangiogenic factors such as vascular endothelial growth factor (VEGF). However, the specific mechanisms underlying these changes in PCOS remain unknown. In this study, we induced the adipogenic differentiation in preadipocyte 3T3-L1 cells and found that adipocyte-derived exosomes promoted proliferation, migration, tube formation, and VEGFA expression in human ovarian microvascular endothelial cells (HOMECs) by delivering miR-30c-5p. Mechanistically, dual luciferase reporter assay demonstrated that miR-30c-5p directly targeted the 3'- untranslated region (UTR) of suppressor of cytokine signaling 3 (SOCS3) mRNA. In addition, adipocyte-derived exosomal miR-30c-5p activated signal transducer and activator of transcription 3 (STAT3)/VEGFA pathway in HOMECs via targeting SOCS3. In vivo experiments indicated that tail vein injection of adipocyte-derived exosomes exacerbated endocrine and metabolic disorders and ovarian angiogenesis in mice with PCOS via miR-30c-5p. Taken together, the study revealed that adipocyte-derived exosomal miR-30c-5p promotes ovarian angiogenesis via the SOCS3/STAT3/VEGFA pathway, thereby participating in the development of PCOS.
Collapse
Affiliation(s)
- Jian Hu
- Reproductive Medical Center, Renmin Hospital of Wuhan University, Wuhan 430060, China; Hubei Clinic Research Center for Assisted Reproductive Technology and Embryonic Development, Wuhan 430060, China
| | - Fangyou Lin
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Yuchen Yin
- Reproductive Medical Center, Renmin Hospital of Wuhan University, Wuhan 430060, China; Hubei Clinic Research Center for Assisted Reproductive Technology and Embryonic Development, Wuhan 430060, China
| | - Yunjie Shang
- Reproductive Medical Center, Renmin Hospital of Wuhan University, Wuhan 430060, China; Hubei Clinic Research Center for Assisted Reproductive Technology and Embryonic Development, Wuhan 430060, China
| | - Zhuoni Xiao
- Reproductive Medical Center, Renmin Hospital of Wuhan University, Wuhan 430060, China; Hubei Clinic Research Center for Assisted Reproductive Technology and Embryonic Development, Wuhan 430060, China.
| | - Wangming Xu
- Reproductive Medical Center, Renmin Hospital of Wuhan University, Wuhan 430060, China; Hubei Clinic Research Center for Assisted Reproductive Technology and Embryonic Development, Wuhan 430060, China.
| |
Collapse
|
13
|
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: 2.0] [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
|
14
|
Wen D, Ren X, Li H, He Y, Hong Y, Cao J, Zheng C, Dong L, Li X. Low expression of RBP4 in the vitreous humour of patients with proliferative diabetic retinopathy who underwent Conbercept intravitreal injection. Exp Eye Res 2022; 225:109197. [PMID: 35932904 DOI: 10.1016/j.exer.2022.109197] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 06/12/2022] [Accepted: 07/18/2022] [Indexed: 12/29/2022]
Abstract
Intravitreal injection of anti-VEGF antibodies has been widely used in the treatment of proliferative diabetic retinopathy (PDR). However, anti-VEGF drugs can exacerbate fibrosis and eventually lead to retinal detachment. To explore proteins closely related to fibrosis, we conducted proteomic analysis of human vitreous humour collected from PDR patients who have or have not intravitreal Conbercept (IVC) injection. Sixteen vitreous humour samples from PDR patients with preoperative IVC and 20 samples from those without preoperative IVC were examined. An immunodepletion kit was used to remove high-abundance vitreous proteins. Conbercept-induced changes were determined using a tandem mass tag-based quantitative proteomic strategy. Enzyme-linked immunosorbent assays were performed to confirm the concentrations of selected proteins and validate the proteomic results. Based on a false discovery rate between 0.05% and -0.05% and a fold-change > 1.5, 97 proteins were altered (49 higher levels and 48 lower levels) in response to IVC. Differentially expressed proteins were found in the extracellular and intracellular regions and were found to be involved in VEGF binding and VEGF-activated receptor activity. Protein-protein interactions indicated associations with fibrosis, neovascularisation and inflammatory signalling pathways. We found the low levels of RBP4 in the vitreous humour of PDR patients with IVC injection, as revealed by ELISA and proteomic profiling. Moreover, RBP4 significantly restored the mitochondrial function of HRMECs induced by AGEs and down regulated the level of glycolysis. Our study is the first to report that RBP4 decreases in the vitreous humour of PDR patients who underwent Conbercept treatment, thereby verifying the role of RBP4 in glucose metabolism. Results provide evidence for the potential mechanism underlying Conbercept-related fibrosis.
Collapse
Affiliation(s)
- Dejia Wen
- Eye Institute and School of Optometry, Tianjin Medical University Eye Hospital, Tianjin, China; Tianjin Branch of National Clinical Research Center for Ocular Disease, Tianjin Key Laboratory of Retinal Functions and Diseases, Tianjin Branch of National Clinical Research Center for Ocular Disease, 300384, Tianjin, China
| | - Xinjun Ren
- Eye Institute and School of Optometry, Tianjin Medical University Eye Hospital, Tianjin, China; Tianjin Branch of National Clinical Research Center for Ocular Disease, Tianjin Key Laboratory of Retinal Functions and Diseases, Tianjin Branch of National Clinical Research Center for Ocular Disease, 300384, Tianjin, China
| | - Hui Li
- Eye Institute and School of Optometry, Tianjin Medical University Eye Hospital, Tianjin, China; Tianjin Branch of National Clinical Research Center for Ocular Disease, Tianjin Key Laboratory of Retinal Functions and Diseases, Tianjin Branch of National Clinical Research Center for Ocular Disease, 300384, Tianjin, China
| | - Ye He
- Eye Institute and School of Optometry, Tianjin Medical University Eye Hospital, Tianjin, China; Tianjin Branch of National Clinical Research Center for Ocular Disease, Tianjin Key Laboratory of Retinal Functions and Diseases, Tianjin Branch of National Clinical Research Center for Ocular Disease, 300384, Tianjin, China
| | - Yaru Hong
- Eye Institute and School of Optometry, Tianjin Medical University Eye Hospital, Tianjin, China; Tianjin Branch of National Clinical Research Center for Ocular Disease, Tianjin Key Laboratory of Retinal Functions and Diseases, Tianjin Branch of National Clinical Research Center for Ocular Disease, 300384, Tianjin, China
| | - Jingjing Cao
- Eye Institute and School of Optometry, Tianjin Medical University Eye Hospital, Tianjin, China; Tianjin Branch of National Clinical Research Center for Ocular Disease, Tianjin Key Laboratory of Retinal Functions and Diseases, Tianjin Branch of National Clinical Research Center for Ocular Disease, 300384, Tianjin, China
| | - Chuanzhen Zheng
- Eye Institute and School of Optometry, Tianjin Medical University Eye Hospital, Tianjin, China; Tianjin Branch of National Clinical Research Center for Ocular Disease, Tianjin Key Laboratory of Retinal Functions and Diseases, Tianjin Branch of National Clinical Research Center for Ocular Disease, 300384, Tianjin, China
| | - Lijie Dong
- Eye Institute and School of Optometry, Tianjin Medical University Eye Hospital, Tianjin, China; Tianjin Branch of National Clinical Research Center for Ocular Disease, Tianjin Key Laboratory of Retinal Functions and Diseases, Tianjin Branch of National Clinical Research Center for Ocular Disease, 300384, Tianjin, China.
| | - Xiaorong Li
- Eye Institute and School of Optometry, Tianjin Medical University Eye Hospital, Tianjin, China; Tianjin Branch of National Clinical Research Center for Ocular Disease, Tianjin Key Laboratory of Retinal Functions and Diseases, Tianjin Branch of National Clinical Research Center for Ocular Disease, 300384, Tianjin, China.
| |
Collapse
|
15
|
Fu Z, Nilsson AK, Hellstrom A, Smith LEH. Retinopathy of prematurity: Metabolic risk factors. eLife 2022; 11:e80550. [PMID: 36420952 PMCID: PMC9691009 DOI: 10.7554/elife.80550] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Accepted: 11/16/2022] [Indexed: 11/25/2022] Open
Abstract
At preterm birth, the retina is incompletely vascularized. Retinopathy of prematurity (ROP) is initiated by the postnatal suppression of physiological retinal vascular development that would normally occur in utero. As the neural retina slowly matures, increasing metabolic demand including in the peripheral avascular retina, leads to signals for compensatory but pathological neovascularization. Currently, only late neovascular ROP is treated. ROP could be prevented by promoting normal vascular growth. Early perinatal metabolic dysregulation is a strong but understudied risk factor for ROP and other long-term sequelae of preterm birth. We will discuss the metabolic and oxygen needs of retina, current treatments, and potential interventions to promote normal vessel growth including control of postnatal hyperglycemia, dyslipidemia and hyperoxia-induced retinal metabolic alterations. Early supplementation of missing nutrients and growth factors and control of supplemental oxygen promotes physiological retinal development. We will discuss the current knowledge gap in retinal metabolism after preterm birth.
Collapse
Affiliation(s)
- Zhongjie Fu
- Department of Ophthalmology, Boston Children’s Hospital, Harvard Medical SchoolBostonUnited States
| | - Anders K Nilsson
- The Sahlgrenska Centre for Pediatric Ophthalmology Research, Department of Clinical Neuroscience, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of GothenburgGothenburgSweden
| | - Ann Hellstrom
- The Sahlgrenska Centre for Pediatric Ophthalmology Research, Department of Clinical Neuroscience, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of GothenburgGothenburgSweden
| | - Lois EH Smith
- Department of Ophthalmology, Boston Children’s Hospital, Harvard Medical SchoolBostonUnited States
| |
Collapse
|
16
|
Neurovascular abnormalities in retinopathy of prematurity and emerging therapies. J Mol Med (Berl) 2022; 100:817-828. [PMID: 35394143 DOI: 10.1007/s00109-022-02195-2] [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: 03/01/2022] [Revised: 03/01/2022] [Accepted: 03/28/2022] [Indexed: 10/18/2022]
Abstract
Blood vessels in the developing retina are formed in concert with neural growth, resulting in functional neurovascular network. Disruption of the neurovascular coordination contributes to the pathogenesis of retinopathy of prematurity (ROP), a potentially blinding retinal neovascular disease in preterm infants that currently lacks an approved drug therapy in the USA. Despite vasculopathy as predominant clinical manifestations, an increasing number of studies revealed complex neurovascular interplays among neurons, glial cells and blood vessels during ROP. Coordinated expression of glia-derived vascular endothelial growth factor (VEGF) in spatio-temporal gradients is pivotal to the formation of well-organized vascular plexuses in the healthy retina, whereas uncoordinated VEGF expression triggers pathological angiogenesis with disorganized vascular tufts in ROP. In contrast with VEGF driving both pathological and physiological angiogenesis, neuron-derived angiogenic factor secretogranin III (Scg3) stringently regulates ROP but not healthy retinal vessels in animal models. Anti-VEGF and anti-Scg3 therapies confer similar high efficacies to alleviate ROP in preclinical studies but are distinct in their disease selectivity and safety. This review discusses neurovascular communication among retinal blood vessels, neurons and glial cells during retinal development and ROP pathogenesis and summarizes the current and emerging therapies to address unmet clinical needs for the disease.
Collapse
|
17
|
Wang T, Zhou P, Xie X, Tomita Y, Cho S, Tsirukis D, Lam E, Luo HR, Sun Y. Myeloid lineage contributes to pathological choroidal neovascularization formation via SOCS3. EBioMedicine 2021; 73:103632. [PMID: 34688035 PMCID: PMC8546367 DOI: 10.1016/j.ebiom.2021.103632] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Revised: 09/27/2021] [Accepted: 10/05/2021] [Indexed: 01/08/2023] Open
Abstract
Background Pathological neovascularization in neovascular age-related macular degeneration (nAMD) is the leading cause of vision loss in the elderly. Increasing evidence shows that cells of myeloid lineage play important roles in controlling pathological endothelium formation. Suppressor of cytokine signaling 3 (SOCS3) pathway has been linked to neovascularization. Methods We utilised a laser-induced choroidal neovascularization (CNV) mouse model to investigate the neovascular aspect of human AMD. In several cell lineage reporter mice, bone marrow chimeric mice and Socs3 loss-of-function (knockout) and gain-of-function (overexpression) mice, immunohistochemistry, confocal, and choroidal explant co-culture with bone marrow-derived macrophage medium were used to study the mechanisms underlying pathological CNV formation via myeloid SOCS3. Findings SOCS3 was significantly induced in myeloid lineage cells, which were recruited into the CNV lesion area. Myeloid Socs3 overexpression inhibited laser-induced CNV, reduced myeloid lineage-derived macrophage/microglia recruitment onsite, and attenuated pro-inflammatory factor expression. Moreover, SOCS3 in myeloid regulated vascular sprouting ex vivo in choroid explants and SOCS3 agonist reduced in vivo CNV. Interpretation These findings suggest that myeloid lineage cells contributed to pathological CNV formation regulated by SOCS3. Funding This project was funded by NIH/NEI (R01EY030140, R01EY029238), BrightFocus Foundation, American Health Assistance Foundation (AHAF), and Boston Children's Hospital Ophthalmology Foundation for YS and the National Institutes of Health/National Heart, Lung and Blood Institute (U01HL098166) for PZ.
Collapse
Affiliation(s)
- Tianxi Wang
- Department of Ophthalmology, Harvard Medical School, Boston Children's Hospital, Boston, MA, USA
| | - Pingzhu Zhou
- Department of Cardiology, Harvard Medical School, Boston Children's Hospital, Boston, MA, USA
| | - Xuemei Xie
- Division of Blood Bank, Department of Laboratory Medicine, Stem Cell Program, Boston Children's Hospital, Boston, MA, USA
| | - Yohei Tomita
- Department of Ophthalmology, Harvard Medical School, Boston Children's Hospital, Boston, MA, USA
| | - Steve Cho
- Department of Ophthalmology, Harvard Medical School, Boston Children's Hospital, Boston, MA, USA
| | - Demetrios Tsirukis
- Department of Ophthalmology, Harvard Medical School, Boston Children's Hospital, Boston, MA, USA
| | - Enton Lam
- Department of Ophthalmology, Harvard Medical School, Boston Children's Hospital, Boston, MA, USA
| | - Hongbo Robert Luo
- Division of Blood Bank, Department of Laboratory Medicine, Stem Cell Program, Boston Children's Hospital, Boston, MA, USA; Joint Program in Transfusion Medicine, Department of Pathology, Harvard Medical School, Boston, MA, USA; Dana-Farber/Harvard Cancer Center, Boston, MA, USA
| | - Ye Sun
- Department of Ophthalmology, Harvard Medical School, Boston Children's Hospital, Boston, MA, USA.
| |
Collapse
|
18
|
Kasembeli MM, Singhmar P, Ma J, Edralin J, Tang Y, Adams C, Heijnen CJ, Kavelaars A, Tweardy DJ. TTI-101: A competitive inhibitor of STAT3 that spares oxidative phosphorylation and reverses mechanical allodynia in mouse models of neuropathic pain. Biochem Pharmacol 2021; 192:114688. [PMID: 34274354 PMCID: PMC8478865 DOI: 10.1016/j.bcp.2021.114688] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Revised: 07/11/2021] [Accepted: 07/13/2021] [Indexed: 01/06/2023]
Abstract
Signal Transducer and Activator of Transcription (STAT) 3 emerged rapidly as a high-value target for treatment of cancer. However, small-molecule STAT3 inhibitors have been slow to enter the clinic due, in part, to serious adverse events (SAE), including lactic acidosis and peripheral neuropathy, which have been attributed to inhibition of STAT3's mitochondrial function. Our group developed TTI-101, a competitive inhibitor of STAT3 that targets the receptor pY705-peptide binding site within the Src homology 2 (SH2) domain to block its recruitment and activation. TTI-101 has shown target engagement, no toxicity, and evidence of clinical benefit in a Phase I study in patients with solid tumors. Here we report that TTI-101 did not affect mitochondrial function, nor did it cause STAT3 aggregation, chemically modify STAT3 or cause neuropathic pain. Instead, TTI-101 unexpectedly suppressed neuropathic pain induced by chemotherapy or in a spared nerve injury model. Thus, in addition to its direct anti-tumor effect, TTI-101 may be of benefit when administered to cancer patients at risk of developing chemotherapy-induced peripheral neuropathy (CIPN).
Collapse
Affiliation(s)
- Moses M Kasembeli
- The Department of Infectious Diseases, Infection Control & Employee Health, Division of Internal Medicine, University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX 77030-4009, United States
| | - Pooja Singhmar
- The Department of Symptom Research, Division of Internal Medicine, University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX 77030-4009, United States
| | - Jiacheng Ma
- The Department of Symptom Research, Division of Internal Medicine, University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX 77030-4009, United States
| | - Jules Edralin
- The Department of Symptom Research, Division of Internal Medicine, University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX 77030-4009, United States
| | - Yongfu Tang
- The Department of Symptom Research, Division of Internal Medicine, University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX 77030-4009, United States
| | - Clydell Adams
- The Department of Infectious Diseases, Infection Control & Employee Health, Division of Internal Medicine, University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX 77030-4009, United States
| | - Cobi J Heijnen
- The Department of Symptom Research, Division of Internal Medicine, University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX 77030-4009, United States
| | - Annemieke Kavelaars
- The Department of Symptom Research, Division of Internal Medicine, University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX 77030-4009, United States
| | - David J Tweardy
- The Department of Infectious Diseases, Infection Control & Employee Health, Division of Internal Medicine, University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX 77030-4009, United States.
| |
Collapse
|
19
|
Gao Y, Liu R, He C, Basile J, Vesterlund M, Wahren-Herlenius M, Espinoza A, Hokka-Zakrisson C, Zadjali F, Yoshimura A, Karlsson M, Carow B, Rottenberg ME. SOCS3 Expression by Thymic Stromal Cells Is Required for Normal T Cell Development. Front Immunol 2021; 12:642173. [PMID: 33815395 PMCID: PMC8012910 DOI: 10.3389/fimmu.2021.642173] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Accepted: 02/22/2021] [Indexed: 12/12/2022] Open
Abstract
The suppressor of cytokine signaling 3 (SOCS3) is a major regulator of immune responses and inflammation as it negatively regulates cytokine signaling. Here, the role of SOCS3 in thymic T cell formation was studied in Socs3fl/flActin-creER mice (Δsocs3) with a tamoxifen inducible and ubiquitous Socs3 deficiency. Δsocs3 thymi showed a 90% loss of cellularity and altered cortico-medullary organization. Thymocyte differentiation and proliferation was impaired at the early double negative (CD4-CD8-) cell stage and apoptosis was increased during the double positive (CD4+CD8+) cell stage, resulting in the reduction of recent thymic emigrants in peripheral organs. Using bone marrow chimeras, transplanting thymic organoids and using mice deficient of SOCS3 in thymocytes we found that expression in thymic stromal cells rather than in thymocytes was critical for T cell development. We found that SOCS3 in thymic epithelial cells (TECs) binds to the E3 ubiquitin ligase TRIM 21 and that Trim21−/− mice showed increased thymic cellularity. Δsocs3 TECs showed alterations in the expression of genes involved in positive and negative selection and lympho-stromal interactions. SOCS3-dependent signal inhibition of the common gp130 subunit of the IL-6 receptor family was redundant for T cell formation. Together, SOCS3 expression in thymic stroma cells is critical for T cell development and for maintenance of thymus architecture.
Collapse
Affiliation(s)
- Yu Gao
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - Ruining Liu
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - Chenfei He
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - Juan Basile
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - Mattias Vesterlund
- SciLife Lab, Department of Oncology-Patohology, Karolinska Institutet, Stockholm, Sweden
| | - Marie Wahren-Herlenius
- Department of Medicine, Karolinska Institutet, Stockholm, Sweden.,Broegelmann Research Laboratory, Department of Clinical Science, University of Bergen, Bergen, Norway
| | | | | | - Fahad Zadjali
- College of Medicine and Health Sciences, Sultan Qaboos University, Muscat, Oman
| | - Akihiko Yoshimura
- Department of Microbiology and Immunology, Keio University School of Medicine, Tokyo, Japan
| | - Mikael Karlsson
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - Berit Carow
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - Martin E Rottenberg
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
20
|
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: 0.8] [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
|
21
|
Hombrebueno JR, Lynch A, Byrne EM, Obasanmi G, Kissenpfennig A, Chen M, Xu H. Hyaloid Vasculature as a Major Source of STAT3 + (Signal Transducer and Activator of Transcription 3) Myeloid Cells for Pathogenic Retinal Neovascularization in Oxygen-Induced Retinopathy. Arterioscler Thromb Vasc Biol 2020; 40:e367-e379. [PMID: 33115265 DOI: 10.1161/atvbaha.120.314567] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
OBJECTIVE Myeloid cells are critically involved in inflammation-induced angiogenesis, although their pathogenic role in the ischemic retina remains controversial. We hypothesize that myeloid cells contribute to pathogenic neovascularization in retinopathy of prematurity through STAT3 (signal transducer and activator of transcription 3) activation. Approach and Results: Using the mouse model of oxygen-induced retinopathy, we show that myeloid cells (CD45+IsolectinB4 [IB4]+) and particularly M2-type macrophages (CD45+ Arg1+), comprise a major source of STAT3 activation (pSTAT3) in the immature ischemic retina. Most of the pSTAT3-expressing myeloid cells concentrated at the hyaloid vasculature and their numbers were strongly correlated with the severity of pathogenic neovascular tuft formation. Pharmacological inhibition of STAT3 reduced the load of IB4+ cells in the hyaloid vasculature and significantly reduced the formation of pathogenic neovascular tufts during oxygen-induced retinopathy, leading to improved long-term visual outcomes (ie, increased retinal thickness and scotopic b-wave electroretinogram responses). Genetic deletion of SOCS3 (suppressor of cytokine signaling 3), an endogenous inhibitor of STAT3, in myeloid cells, enhanced pathological and physiological neovascularization in oxygen-induced retinopathy, indicating that myeloid-STAT3 signaling is crucial for retinal angiogenesis. CONCLUSIONS Circulating myeloid cells may migrate to the immature ischemic retina through the hyaloid vasculature and contribute to retinal neovascularization via activation of STAT3. Understanding how STAT3 modulates myeloid cells for vascular repair/pathology may provide novel therapeutic options in pathogenic angiogenesis.
Collapse
Affiliation(s)
- Jose R Hombrebueno
- Wellcome-Wolfson Institute for Experimental Medicine, School of Medicine, Dentistry and Biomedical Sciences, Queen's University Belfast, United Kingdom (J.R.H., A.L., E.M.B., G.O., A.K., M.C., H.X.).,Institute of Inflammation and Ageing, College of Medical and Dental Sciences, University of Birmingham, United Kingdom (J.R.H.)
| | - Aisling Lynch
- Wellcome-Wolfson Institute for Experimental Medicine, School of Medicine, Dentistry and Biomedical Sciences, Queen's University Belfast, United Kingdom (J.R.H., A.L., E.M.B., G.O., A.K., M.C., H.X.)
| | - Eimear M Byrne
- Wellcome-Wolfson Institute for Experimental Medicine, School of Medicine, Dentistry and Biomedical Sciences, Queen's University Belfast, United Kingdom (J.R.H., A.L., E.M.B., G.O., A.K., M.C., H.X.)
| | - Gideon Obasanmi
- Wellcome-Wolfson Institute for Experimental Medicine, School of Medicine, Dentistry and Biomedical Sciences, Queen's University Belfast, United Kingdom (J.R.H., A.L., E.M.B., G.O., A.K., M.C., H.X.)
| | - Adrien Kissenpfennig
- Wellcome-Wolfson Institute for Experimental Medicine, School of Medicine, Dentistry and Biomedical Sciences, Queen's University Belfast, United Kingdom (J.R.H., A.L., E.M.B., G.O., A.K., M.C., H.X.)
| | - Mei Chen
- Wellcome-Wolfson Institute for Experimental Medicine, School of Medicine, Dentistry and Biomedical Sciences, Queen's University Belfast, United Kingdom (J.R.H., A.L., E.M.B., G.O., A.K., M.C., H.X.)
| | - Heping Xu
- Wellcome-Wolfson Institute for Experimental Medicine, School of Medicine, Dentistry and Biomedical Sciences, Queen's University Belfast, United Kingdom (J.R.H., A.L., E.M.B., G.O., A.K., M.C., H.X.)
| |
Collapse
|
22
|
Wang T, Tsirukis DI, Sun Y. Targeting Neuroinflammation in Neovascular Retinal Diseases. Front Pharmacol 2020; 11:234. [PMID: 32210818 PMCID: PMC7076162 DOI: 10.3389/fphar.2020.00234] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2019] [Accepted: 02/20/2020] [Indexed: 12/12/2022] Open
Abstract
Retinal blood vessels provide the necessary energy, nutrients and oxygen in order to support visual function and remove harmful particles from blood, thus acting to protect neuronal cells. The homeostasis of the retinal vessels is important for the maintenance of retinal visual function. Neovascularization is the most common cause of blindness in patients with retinopathy. Previous studies have shown that inflammatory mediators are known key regulators in retinopathy, but their causal link has been elusive. Although inflammation is often thought to arise from inflammatory cells like macrophages, neutrophils, and resident microglia, retinal neurons have also been reported to contribute to inflammation, through inflammatory signals, which mediate blood vessel growth. Therefore, it is important to explore the detailed mechanisms of neuroinflammation’s effects on retinal neovascularization. This review looks to summarize current research on the relationship between retinal angiogenesis and neuroinflammation in retinopathy, as well as the potential effects of neuroinflammation on retinal neovascularization in different animal models.
Collapse
Affiliation(s)
- Tianxi Wang
- Department of Ophthalmology, Boston Children's Hospital, Harvard Medical School, Boston, MA, United States
| | - Demetrios I Tsirukis
- Department of Ophthalmology, Boston Children's Hospital, Harvard Medical School, Boston, MA, United States
| | - Ye Sun
- Department of Ophthalmology, Boston Children's Hospital, Harvard Medical School, Boston, MA, United States
| |
Collapse
|
23
|
Fu Z, Sun Y, Cakir B, Tomita Y, Huang S, Wang Z, Liu CH, S. Cho S, Britton W, S. Kern T, Antonetti DA, Hellström A, E.H. Smith L. Targeting Neurovascular Interaction in Retinal Disorders. Int J Mol Sci 2020; 21:E1503. [PMID: 32098361 PMCID: PMC7073081 DOI: 10.3390/ijms21041503] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2020] [Revised: 02/13/2020] [Accepted: 02/21/2020] [Indexed: 02/07/2023] Open
Abstract
The tightly structured neural retina has a unique vascular network comprised of three interconnected plexuses in the inner retina (and choroid for outer retina), which provide oxygen and nutrients to neurons to maintain normal function. Clinical and experimental evidence suggests that neuronal metabolic needs control both normal retinal vascular development and pathological aberrant vascular growth. Particularly, photoreceptors, with the highest density of mitochondria in the body, regulate retinal vascular development by modulating angiogenic and inflammatory factors. Photoreceptor metabolic dysfunction, oxidative stress, and inflammation may cause adaptive but ultimately pathological retinal vascular responses, leading to blindness. Here we focus on the factors involved in neurovascular interactions, which are potential therapeutic targets to decrease energy demand and/or to increase energy production for neovascular retinal disorders.
Collapse
Affiliation(s)
- Zhongjie Fu
- Department of Ophthalmology, Boston Children’s Hospital, Harvard Medical School, Boston, MA 02115, USA; (Z.F.); (Y.S.); (B.C.); (Y.T.); (S.H.); (Z.W.); (C.-H.L.); (S.S.C.); (W.B.)
- Manton Center for Orphan Disease, Boston Children’s Hospital, Boston, MA 02115, USA
| | - Ye Sun
- Department of Ophthalmology, Boston Children’s Hospital, Harvard Medical School, Boston, MA 02115, USA; (Z.F.); (Y.S.); (B.C.); (Y.T.); (S.H.); (Z.W.); (C.-H.L.); (S.S.C.); (W.B.)
| | - Bertan Cakir
- Department of Ophthalmology, Boston Children’s Hospital, Harvard Medical School, Boston, MA 02115, USA; (Z.F.); (Y.S.); (B.C.); (Y.T.); (S.H.); (Z.W.); (C.-H.L.); (S.S.C.); (W.B.)
| | - Yohei Tomita
- Department of Ophthalmology, Boston Children’s Hospital, Harvard Medical School, Boston, MA 02115, USA; (Z.F.); (Y.S.); (B.C.); (Y.T.); (S.H.); (Z.W.); (C.-H.L.); (S.S.C.); (W.B.)
| | - Shuo Huang
- Department of Ophthalmology, Boston Children’s Hospital, Harvard Medical School, Boston, MA 02115, USA; (Z.F.); (Y.S.); (B.C.); (Y.T.); (S.H.); (Z.W.); (C.-H.L.); (S.S.C.); (W.B.)
| | - Zhongxiao Wang
- Department of Ophthalmology, Boston Children’s Hospital, Harvard Medical School, Boston, MA 02115, USA; (Z.F.); (Y.S.); (B.C.); (Y.T.); (S.H.); (Z.W.); (C.-H.L.); (S.S.C.); (W.B.)
| | - Chi-Hsiu Liu
- Department of Ophthalmology, Boston Children’s Hospital, Harvard Medical School, Boston, MA 02115, USA; (Z.F.); (Y.S.); (B.C.); (Y.T.); (S.H.); (Z.W.); (C.-H.L.); (S.S.C.); (W.B.)
| | - Steve S. Cho
- Department of Ophthalmology, Boston Children’s Hospital, Harvard Medical School, Boston, MA 02115, USA; (Z.F.); (Y.S.); (B.C.); (Y.T.); (S.H.); (Z.W.); (C.-H.L.); (S.S.C.); (W.B.)
| | - William Britton
- Department of Ophthalmology, Boston Children’s Hospital, Harvard Medical School, Boston, MA 02115, USA; (Z.F.); (Y.S.); (B.C.); (Y.T.); (S.H.); (Z.W.); (C.-H.L.); (S.S.C.); (W.B.)
| | - Timothy S. Kern
- Center for Translational Vision Research, Gavin Herbert Eye Institute, Irvine, CA 92697, USA;
| | - David A. Antonetti
- Kellogg Eye Center, Department of Ophthalmology and Visual Sciences, University of Michigan, Ann Arbor, MI 48105, USA;
| | - Ann Hellström
- Section for Ophthalmology, Department of Clinical Neuroscience, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, 405 30 Göteborg, Sweden;
| | - Lois E.H. Smith
- Department of Ophthalmology, Boston Children’s Hospital, Harvard Medical School, Boston, MA 02115, USA; (Z.F.); (Y.S.); (B.C.); (Y.T.); (S.H.); (Z.W.); (C.-H.L.); (S.S.C.); (W.B.)
| |
Collapse
|
24
|
Yang H, Gan S, Jiang Z, Song X, Chen T, Xu Y, Fu L, Zhang Y, Tao L, Shen X. Protective effects of essential oil from Fructus Alpiniae zerumbet on retinal Müller gliosis via the PPAR-γ-p-CREB signaling pathway. Chin Med 2020; 15:4. [PMID: 31938037 PMCID: PMC6954544 DOI: 10.1186/s13020-019-0283-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2019] [Accepted: 12/27/2019] [Indexed: 12/18/2022] Open
Abstract
Background Diabetic retinopathy (DR) involves extensive retinal damage and is one of the most common and serious complications of diabetes mellitus. Hyperglycemia is the major pathological trigger for diabetic complications. Müller cell gliosis, a key pathophysiological process in DR, could finally lead to vision loss. Our previous finding revealed that the essential oil of Fructus Alpiniae zerumbet (EOFAZ) protects human umbilical vein endothelial cells (HUVECs) against high glucose (HG)-induced injury via the PPAR-γ signal. However, Whether EOFAZ could prevent HG-induced Müller cell gliosis through the PPAR signaling remains unclear. Methods The neuroprotective effects of EOFAZ were evaluated in HG-treated rat retinal Müller cells (RMCs) and DR rat model. Result GFAP and VEGF upregulation is the biomarker of Müller glial reactivity gliosis. Results suggested that EOFAZ could remarkably ameliorate retinal reactive gliosis by suppressing p-CREB and GFAP and VEGF downstream effectors. Its effects on PPAR-γ, a major target for currently available anti-diabetes drugs, were also investigated. EOFAZ treatment remarkably attenuated the reduction of PPAR-γ and high level of p-CaMK II and p-CREB in HG-treated RMCs and diabetic rats. Furthermore, the activation and ectopic expression of PPAR-γ downregulated p-CREB and p-CaMK II in HG-treated RMCs. By contrast, CaMK II inhibitor KN93 and CREB gene silencing did not significantly affect the PPAR-γ expression. Conclusions A novel PPAR-γ-p-CREB signaling pathway accounts for the inhibitory effect of EOFAZ on RMCs gliosis. These findings provide scientific evidence for the potential use of EOFAZ as a complementary and alternative medicine for DR prevention and treatment in the future.
Collapse
Affiliation(s)
- Hong Yang
- 1The Department of Pharmacology of Materia Medica (the State Key Laboratory of Functions and Applications of Medicinal Plants, the High Educational Key Laboratory of Guizhou Province for Natural Medicinal Pharmacology and Druggability), School of Pharmaceutical Sciences, Guizhou Medical University, University Town, Guian New District, Guizhou, 550025 China.,2The High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou Province (the Union Key Laboratory of Guiyang City-Guizhou Medical University), School of Pharmaceutical Sciences, Guizhou Medical University, University Town, Guian New District, Guizhou, China
| | - Shiquan Gan
- 1The Department of Pharmacology of Materia Medica (the State Key Laboratory of Functions and Applications of Medicinal Plants, the High Educational Key Laboratory of Guizhou Province for Natural Medicinal Pharmacology and Druggability), School of Pharmaceutical Sciences, Guizhou Medical University, University Town, Guian New District, Guizhou, 550025 China.,2The High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou Province (the Union Key Laboratory of Guiyang City-Guizhou Medical University), School of Pharmaceutical Sciences, Guizhou Medical University, University Town, Guian New District, Guizhou, China
| | - Zhaohui Jiang
- 1The Department of Pharmacology of Materia Medica (the State Key Laboratory of Functions and Applications of Medicinal Plants, the High Educational Key Laboratory of Guizhou Province for Natural Medicinal Pharmacology and Druggability), School of Pharmaceutical Sciences, Guizhou Medical University, University Town, Guian New District, Guizhou, 550025 China.,2The High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou Province (the Union Key Laboratory of Guiyang City-Guizhou Medical University), School of Pharmaceutical Sciences, Guizhou Medical University, University Town, Guian New District, Guizhou, China
| | - Xiaomei Song
- 1The Department of Pharmacology of Materia Medica (the State Key Laboratory of Functions and Applications of Medicinal Plants, the High Educational Key Laboratory of Guizhou Province for Natural Medicinal Pharmacology and Druggability), School of Pharmaceutical Sciences, Guizhou Medical University, University Town, Guian New District, Guizhou, 550025 China.,2The High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou Province (the Union Key Laboratory of Guiyang City-Guizhou Medical University), School of Pharmaceutical Sciences, Guizhou Medical University, University Town, Guian New District, Guizhou, China
| | - Tingting Chen
- 1The Department of Pharmacology of Materia Medica (the State Key Laboratory of Functions and Applications of Medicinal Plants, the High Educational Key Laboratory of Guizhou Province for Natural Medicinal Pharmacology and Druggability), School of Pharmaceutical Sciences, Guizhou Medical University, University Town, Guian New District, Guizhou, 550025 China.,2The High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou Province (the Union Key Laboratory of Guiyang City-Guizhou Medical University), School of Pharmaceutical Sciences, Guizhou Medical University, University Town, Guian New District, Guizhou, China.,3The Key Laboratory of Optimal Utilization of Natural Medicine Resources, School of Pharmaceutical Sciences, Guizhou Medical University, University Town, Guian New District, Guizhou, China
| | - Yini Xu
- 1The Department of Pharmacology of Materia Medica (the State Key Laboratory of Functions and Applications of Medicinal Plants, the High Educational Key Laboratory of Guizhou Province for Natural Medicinal Pharmacology and Druggability), School of Pharmaceutical Sciences, Guizhou Medical University, University Town, Guian New District, Guizhou, 550025 China.,2The High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou Province (the Union Key Laboratory of Guiyang City-Guizhou Medical University), School of Pharmaceutical Sciences, Guizhou Medical University, University Town, Guian New District, Guizhou, China
| | - Lingyun Fu
- 2The High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou Province (the Union Key Laboratory of Guiyang City-Guizhou Medical University), School of Pharmaceutical Sciences, Guizhou Medical University, University Town, Guian New District, Guizhou, China.,3The Key Laboratory of Optimal Utilization of Natural Medicine Resources, School of Pharmaceutical Sciences, Guizhou Medical University, University Town, Guian New District, Guizhou, China
| | - Yanyan Zhang
- 1The Department of Pharmacology of Materia Medica (the State Key Laboratory of Functions and Applications of Medicinal Plants, the High Educational Key Laboratory of Guizhou Province for Natural Medicinal Pharmacology and Druggability), School of Pharmaceutical Sciences, Guizhou Medical University, University Town, Guian New District, Guizhou, 550025 China.,2The High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou Province (the Union Key Laboratory of Guiyang City-Guizhou Medical University), School of Pharmaceutical Sciences, Guizhou Medical University, University Town, Guian New District, Guizhou, China
| | - Ling Tao
- 3The Key Laboratory of Optimal Utilization of Natural Medicine Resources, School of Pharmaceutical Sciences, Guizhou Medical University, University Town, Guian New District, Guizhou, China
| | - Xiangchun Shen
- 1The Department of Pharmacology of Materia Medica (the State Key Laboratory of Functions and Applications of Medicinal Plants, the High Educational Key Laboratory of Guizhou Province for Natural Medicinal Pharmacology and Druggability), School of Pharmaceutical Sciences, Guizhou Medical University, University Town, Guian New District, Guizhou, 550025 China.,2The High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou Province (the Union Key Laboratory of Guiyang City-Guizhou Medical University), School of Pharmaceutical Sciences, Guizhou Medical University, University Town, Guian New District, Guizhou, China.,3The Key Laboratory of Optimal Utilization of Natural Medicine Resources, School of Pharmaceutical Sciences, Guizhou Medical University, University Town, Guian New District, Guizhou, China
| |
Collapse
|
25
|
Xu CH, Liu Y, Xiao LM, Chen LK, Zheng SY, Zeng EM, Li DH, Li YP. Silencing microRNA-221/222 cluster suppresses glioblastoma angiogenesis by suppressor of cytokine signaling-3-dependent JAK/STAT pathway. J Cell Physiol 2019; 234:22272-22284. [PMID: 31106423 DOI: 10.1002/jcp.28794] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2019] [Revised: 04/17/2019] [Accepted: 04/22/2019] [Indexed: 12/13/2022]
Abstract
Angiogenesis is a major pathologic characteristic of glioblastoma, which is one aggressive primary brain tumor. MicroRNA-221/222 (miR-221/222) cluster has been previously reported to function importantly in malignant glioma biological process. The current study aims at evaluating the effects of miR-221/222 cluster on angiogenesis of glioblastoma cells. Microarray data were analyzed to select glioblastoma-associated differentially expressed genes, and dual-luciferase reporter assay was performed to assess targeting correlation between miR-221/222 cluster and suppressor of cytokine signaling-3 (SOCS3). Subsequently, the expression patterns of miR-221 and miR-222 in glioblastoma cells were identified. miR-221 and miR-222 were overexpressed or silenced in glioblastoma cells to identify the effect of miR-221/222 cluster in cell invasion, migration, proliferation, and angiogenesis. To define downstream pathway of miR-221/222 cluster or SOCS3 in glioblastoma, levels of Janus kinase (JAK)/signal transducers and activators of transcription (STAT) pathway-related proteins were assessed. Additionally, the functions of miR-221/222 on glioblastoma cell angiogenesis were measured in vivo with microvessel density assayed. miR-221 and miR-222 were expressed at a high level and SOCS3 was at a low level in glioblastoma. Downregulation of the miR-221/222 cluster diminished the invasion, migration, proliferation, and angiogenesis with reduced protein levels of matrix metalloproteinase-2 (MMP-2), MMP-9, and vascular endothelial growth factor in glioblastoma cells. Also, silencing miR-221/222 cluster reduced p-JAK2/JAK2 and p-STAT3/STAT3. Consistently, the inhibitory role of silencing miR-221/222 cluster on tumorigenesis of glioblastoma cells was confirmed in vivo. Collectively, the inhibition of miR-221/222 cluster could attenuate the glioblastoma angiogenesis through inactivation of the JAK/STAT pathway by upregulating SOCS3.
Collapse
Affiliation(s)
- Chun-Hua Xu
- Department of Neurosurgery, The First Affiliated Hospital of Nanchang University, Nanchang, P.R. China
| | - Yue Liu
- Department of Neurosurgery, The First Affiliated Hospital of Nanchang University, Nanchang, P.R. China
| | - Li-Min Xiao
- Department of Neurosurgery, The First Affiliated Hospital of Nanchang University, Nanchang, P.R. China
| | - Li-Ke Chen
- Department of Neurosurgery, The First Affiliated Hospital of Nanchang University, Nanchang, P.R. China
| | - Su-Yue Zheng
- Department of Neurosurgery, The First Affiliated Hospital of Nanchang University, Nanchang, P.R. China
| | - Er-Ming Zeng
- Department of Neurosurgery, The First Affiliated Hospital of Nanchang University, Nanchang, P.R. China
| | - Dong-Hai Li
- Department of Neurosurgery, The First Affiliated Hospital of Nanchang University, Nanchang, P.R. China
| | - You-Ping Li
- Department of Neurosurgery, The First Affiliated Hospital of Nanchang University, Nanchang, P.R. China
| |
Collapse
|
26
|
Ock S, Park S, Lee J, Kim J. RANKL blockade suppresses pathological angiogenesis and vascular leakage in ischemic retinopathy. Biochem Biophys Res Commun 2019; 516:350-356. [PMID: 31208720 DOI: 10.1016/j.bbrc.2019.06.046] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2019] [Accepted: 06/09/2019] [Indexed: 11/27/2022]
Abstract
Receptor activator of NF-κB ligand (RANKL) is a member of the TNF superfamily. RANKL increases endothelial permeability and induces angiogenesis, suggesting its critical roles in the vasculature. Despite the evidence implicating RANKL in vascular pathology, its role in ischemic retinopathy has not been previously reported. In this study, neonatal mice were exposed to 75% oxygen from postnatal day (P)7 to P12 to induce vaso-obliteration, and then returned to room air from P12 to P17, causing the retina to become hypoxic and inducing vascular endothelial growth factor (VEGF) signaling, which produces pathological neovascularization. On P12, the mice received a single intravitreal injection of control IgG1 or RANK-Fc, and retinas were obtained at P17. On P17, RANKL was expressed strongly and selectively in the neovascular tufts (NVT) area. RANKL colocalized with αSMA or PDGFRβ in NVT. However, co-immunostaining revealed that CD31-positive areas were not the same as RANKL, which indicates that RANKL might be produced by retinal pericytes, not endothelial cells. Consistent with this finding, chemical hypoxia upregulated RANKL expression in cultured human retinal pericytes but not in endothelial cells. Treatment with RANK-Fc markedly reduced the NVT area compared to that in mice administered the IgG1 injection. In contrast, the central avascular region of RANKL-Fc retina was comparable to the controls. In addition, we assessed retinal vascular permeability using FITC-labeled dextran. RANK-Fc treated mice displayed decreased vascular leakages compared to those injected with IgG1. Our work supports the use of an RANKL blockade as a potential therapeutic approach against ischemic retinopathies.
Collapse
Affiliation(s)
- Sangmi Ock
- Division of Endocrinology and Metabolism, Department of Internal Medicine, College of Medicine, Chung-Ang University, Seoul, Republic of Korea
| | - Soyoung Park
- Division of Endocrinology and Metabolism, Department of Internal Medicine, College of Medicine, Chung-Ang University, Seoul, Republic of Korea
| | - Junyeop Lee
- Department of Ophthalmology, College of Medicine, Yeungnam University, Daegu, Republic of Korea
| | - Jaetaek Kim
- Division of Endocrinology and Metabolism, Department of Internal Medicine, College of Medicine, Chung-Ang University, Seoul, Republic of Korea.
| |
Collapse
|
27
|
Kuo K, Roberts VHJ, Gaffney J, Takahashi DL, Morgan T, Lo JO, Stouffer RL, Frias AE. Maternal High-Fat Diet Consumption and Chronic Hyperandrogenemia Are Associated With Placental Dysfunction in Female Rhesus Macaques. Endocrinology 2019; 160:1937-1949. [PMID: 31180495 PMCID: PMC6656425 DOI: 10.1210/en.2019-00149] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/24/2019] [Accepted: 06/04/2019] [Indexed: 01/25/2023]
Abstract
The risk of adverse perinatal outcomes with maternal polycystic ovary syndrome may differ among hyperandrogenic and nonhyperandrogenic phenotypes and is likely modulated by maternal obesity and diet. The relative contribution of maternal hyperandrogenism and nutritional status to placental dysfunction is unknown. Female rhesus macaques (N = 39) were assigned at puberty to one of four treatment groups: subcutaneous cholesterol implants and a standard chow diet (controls); testosterone (T) implants and a normal diet; cholesterol implants and a high-fat, Western-style diet (WSD); and testosterone implants in combination with a high-fat diet. After 3.5 years of treatment, contrast-enhanced and Doppler ultrasound analyses of placental blood flow were performed for a representative subset of animals from each treatment group during pregnancy, and placental architecture assessed with stereological analysis. Placental growth factors, cellular nutrient sensors, and angiogenic markers were measured with ELISA and Western blotting. WSD consumption was associated with a 30% increase in placental flux rate relative to that in animals receiving a normal diet. T and WSD treatments were each independently associated with increased villous volume, and T also was associated with an ∼ 40% decrease fetal capillary volume on stereological analysis. T treatment was associated with significantly increased mTOR and SOCS3 expression. WSD consumption was associated with decreased GLUT1 expression and microvillous membrane localization. Hyperandrogenemic and nonhyperandrogenemic phenotypes are associated with altered placental angiogenesis, nutrient sensing, and glucose transport. WSD and T appear to have distinct effects on vascular impedance and capillary angiogenesis.
Collapse
Affiliation(s)
- Kelly Kuo
- Department of Obstetrics & Gynecology, Oregon Health & Science University, Portland, Oregon
- Correspondence: Kelly Kuo, MD, Division of Maternal Fetal Medicine, Department of Obstetrics & Gynecology, Oregon Health & Science University, 3181 Southwest Sam Jackson Park Road, SJH 2356, Portland, Oregon 97239. E-mail:
| | - Victoria H J Roberts
- Division of Reproductive and Developmental Sciences, Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, Oregon
| | - Jessica Gaffney
- Division of Reproductive and Developmental Sciences, Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, Oregon
| | - Diana L Takahashi
- Division of Reproductive and Developmental Sciences, Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, Oregon
| | - Terry Morgan
- Department of Pathology, Oregon Health & Science University, Portland, Oregon
| | - Jamie O Lo
- Department of Obstetrics & Gynecology, Oregon Health & Science University, Portland, Oregon
| | - Richard L Stouffer
- Division of Reproductive and Developmental Sciences, Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, Oregon
| | - Antonio E Frias
- Department of Obstetrics & Gynecology, Oregon Health & Science University, Portland, Oregon
- Division of Reproductive and Developmental Sciences, Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, Oregon
| |
Collapse
|
28
|
Abstract
The retina is one of the most metabolically active tissues in the body, consuming high levels of oxygen and nutrients. A well-organized ocular vascular system adapts to meet the metabolic requirements of the retina to ensure visual function. Pathological conditions affect growth of the blood vessels in the eye. Understanding the neuronal biological processes that govern retinal vascular development is of interest for translational researchers and clinicians to develop preventive and interventional therapeutics for vascular eye diseases that address early drivers of abnormal vascular growth. This review summarizes the current knowledge of the cellular and molecular processes governing both physiological and pathological retinal vascular development, which is dependent on the interaction among retinal cell populations, including neurons, glia, immune cells, and vascular endothelial cells. We also review animal models currently used for studying retinal vascular development.
Collapse
Affiliation(s)
- Ye Sun
- Department of Ophthalmology, Harvard Medical School, Boston Children's Hospital, Boston, Massachusetts 02115, USA;
| | - Lois E H Smith
- Department of Ophthalmology, Harvard Medical School, Boston Children's Hospital, Boston, Massachusetts 02115, USA;
| |
Collapse
|
29
|
m 6A methylation controls pluripotency of porcine induced pluripotent stem cells by targeting SOCS3/JAK2/STAT3 pathway in a YTHDF1/YTHDF2-orchestrated manner. Cell Death Dis 2019; 10:171. [PMID: 30787270 PMCID: PMC6382841 DOI: 10.1038/s41419-019-1417-4] [Citation(s) in RCA: 62] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2018] [Revised: 01/25/2019] [Accepted: 01/28/2019] [Indexed: 12/18/2022]
Abstract
Embryonic stem cells (ESCs) and induced pluripotent stem cells (iPSCs) hold great promise for regenerative medicine, disease treatment, and organ transplantation. As the ethical issue of human ESCs and similarity of pig in human genome and physiological characteristics, the porcine iPSCs (piPSCs) have become an ideal alternative study model. N6-methyladenosine (m6A) methylation is the most prevalent modification in eukaryotic mRNAs, regulating the self-renewal and differentiation of pluripotency stem cells. However, the explicit m6A-regulating machinery remains controversial. Here, we demonstrate that m6A modification and its modulators play a crucial role in mediating piPSCs pluripotency. In brief, loss of METTL3 significantly impairs self-renewal and triggers differentiation of piPSCs by interfering JAK2 and SOCS3 expression, further inactivating JAK2-STAT3 pathway, which then blocks the transcription of KLF4 and SOX2. We identify that both of JAK2 and SOSC3 have m6A modification at 3'UTR by m6A-seq analysis. Dual-luciferase assay shows that METTL3 regulates JAK2 and SOCS3 expression in an m6A-dependent way. RIP-qPCR validates JAK2 and SOCS3 are the targets of YTHDF1 and YTHDF2, respectively. SiMETTL3 induced lower m6A levels of JAK2 and SOCS3 lead to the inhibition of YTHDF1-mediated JAK2 translation and the block of YTHDF2-dependent SOCS3 mRNA decay. Subsequently, the altered protein expressions of JAK2 and SOCS3 inhibit JAK2-STAT3 pathway and then the pluripotency of piPSCs. Collectively, our work uncovers the critical role of m6A modification and its modulators in regulating piPSCs pluripotency and provides insight into an orchestrated network linking the m6A methylation and SOCS3/JAK2/STAT3 pathway in pluripotency regulation.
Collapse
|
30
|
Joly S, Dejda A, Rodriguez L, Sapieha P, Pernet V. Nogo-A inhibits vascular regeneration in ischemic retinopathy. Glia 2018; 66:2079-2093. [PMID: 30051920 DOI: 10.1002/glia.23462] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2017] [Revised: 05/10/2018] [Accepted: 05/10/2018] [Indexed: 01/20/2023]
Abstract
Nogo-A is a potent glial-derived inhibitor of axon growth in the injured CNS and acts as a negative regulator of developmental angiogenesis by inhibiting vascular endothelial cell migration. However, its function in pathological angiogenesis has never been studied after ischemic injury in the CNS. Using the mouse model of oxygen-induced retinopathy (OIR) which yields defined zones of retinal ischemia, our goal was to investigate the role of Nogo-A in vascular regeneration. We demonstrate a marked upregulation of the Nogo-A receptor sphingosine 1-phosphate receptor 2 in blood vessels following OIR, while Nogo-A is abundantly expressed in surrounding glial cells. Acute inhibition of Nogo-A with function-blocking antibody 11C7 significantly improved vascular regeneration and consequently prevented pathological pre-retinal angiogenesis. Ultimately, inhibition of Nogo-A led to restoration of retinal function as determined by electrophysiological response of retinal cells to light stimulation. Our data suggest that anti-Nogo-A antibody may protect neuronal cells from ischemic damage by accelerating blood vessel repair in the CNS. Targeting Nogo-A by immunotherapy may improve CNS perfusion after vascular injuries.
Collapse
Affiliation(s)
- Sandrine Joly
- CUO-Recherche, Centre de recherche du CHU de Québec and Département d'ophtalmologie, Faculté de médecine, Université Laval, Quebec, Quebec, Canada
| | - Agnieszka Dejda
- Department of Ophthalmology, Maisonneuve-Rosemont Hospital Research Centre, University of Montreal, Montreal, Quebec, Canada
| | - Léa Rodriguez
- CUO-Recherche, Centre de recherche du CHU de Québec and Département d'ophtalmologie, Faculté de médecine, Université Laval, Quebec, Quebec, Canada
| | - Przemyslaw Sapieha
- Department of Ophthalmology, Maisonneuve-Rosemont Hospital Research Centre, University of Montreal, Montreal, Quebec, Canada
| | - Vincent Pernet
- CUO-Recherche, Centre de recherche du CHU de Québec and Département d'ophtalmologie, Faculté de médecine, Université Laval, Quebec, Quebec, Canada
| |
Collapse
|
31
|
Xu Y, Lu X, Hu Y, Yang B, Tsui CK, Yu S, Lu L, Liang X. Melatonin attenuated retinal neovascularization and neuroglial dysfunction by inhibition of HIF-1α-VEGF pathway in oxygen-induced retinopathy mice. J Pineal Res 2018; 64:e12473. [PMID: 29411894 DOI: 10.1111/jpi.12473] [Citation(s) in RCA: 83] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/24/2017] [Accepted: 01/25/2018] [Indexed: 12/15/2022]
Abstract
Retinopathy of prematurity (ROP) is a retinopathy characterized by retinal neovascularization (RNV) occurring in preterm infants treated with high concentrations of oxygen and may lead to blindness in severe cases. Currently, anti-VEGF therapy is a major treatment for ROP, but it is costly and may cause serious complications. The previous study has demonstrated that melatonin exerted neuroprotective effect against retinal ganglion cell death induced by hypoxia in neonatal rats. However, whether melatonin is anti-angiogenic and neuroglial protective in the progression of ROP remains unknown. Thus, this study was to investigate the effect of melatonin on RNV and neuroglia in the retina of oxygen-induced retinopathy (OIR) mice. The results showed a reduction in retinal vascular leakage in OIR mice after melatonin treatment. Besides, the size of retinal neovascular and avascular areas, the number of preretinal neovascular cell nuclei, and the number of proliferative vascular endothelial cells within the neovascular area were significantly decreased in mice treated with melatonin. After oxygen-induced injury, the density of astrocytes was decreased, accompanied by morphologic and functional changes of astrocytes. Besides, retinal microglia were also activated. Meanwhile, the levels of inflammatory factors were elevated. However, these pathologic processes were all hindered by melatonin treatment. Furthermore, HIF-1α-VEGF pathway was activated in the retina of OIR mice, yet was suppressed in melatonin-treated OIR mice retinas. In conclusion, melatonin prevented pathologic neovascularization, protected neuroglial cells, and exerts anti-inflammation effect via inhibition of HIF-1α-VEGF pathway in OIR retinas, suggesting that melatonin could be a promising therapeutic agent for ROP.
Collapse
Affiliation(s)
- Yue Xu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, Guangdong Province, China
| | - Xi Lu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, Guangdong Province, China
| | - Yaguang Hu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, Guangdong Province, China
| | - Boyu Yang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, Guangdong Province, China
| | - Ching-Kit Tsui
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, Guangdong Province, China
| | - Shanshan Yu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, Guangdong Province, China
| | - Lin Lu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, Guangdong Province, China
| | - Xiaoling Liang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, Guangdong Province, China
| |
Collapse
|
32
|
Mishra M, Duraisamy AJ, Kowluru RA. Sirt1: A Guardian of the Development of Diabetic Retinopathy. Diabetes 2018; 67:745-754. [PMID: 29311218 PMCID: PMC5860853 DOI: 10.2337/db17-0996] [Citation(s) in RCA: 90] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/21/2017] [Accepted: 12/29/2017] [Indexed: 12/11/2022]
Abstract
Diabetic retinopathy is a multifactorial disease, and the exact mechanism of its pathogenesis remains obscure. Sirtuin 1 (Sirt1), a multifunctional deacetylase, is implicated in the regulation of many cellular functions and in gene transcription, and retinal Sirt1 is inhibited in diabetes. Our aim was to determine the role of Sirt1 in the development of diabetic retinopathy and to elucidate the molecular mechanism of its downregulation. Using Sirt1-overexpressing mice that were diabetic for 8 months, structural, functional, and metabolic abnormalities were investigated in vascular and neuronal retina. The role of epigenetics in Sirt1 transcriptional suppression was investigated in retinal microvessels. Compared with diabetic wild-type mice, retinal vasculature from diabetic Sirt1 mice did not present any increase in the number of apoptotic cells or degenerative capillaries or decrease in vascular density. Diabetic Sirt1 mice were also protected from mitochondrial damage and had normal electroretinography responses and ganglion cell layer thickness. Diabetic wild-type mice had hypermethylated Sirt1 promoter DNA, which was alleviated in diabetic Sirt1 mice, suggesting a role for epigenetics in its transcriptional suppression. Thus strategies targeted to ameliorate Sirt1 inhibition have the potential to maintain retinal vascular and neuronal homeostasis, providing opportunities to retard the development of diabetic retinopathy in its early stages.
Collapse
Affiliation(s)
- Manish Mishra
- Kresge Eye Institute, Wayne State University, Detroit, MI
| | | | - Renu A Kowluru
- Kresge Eye Institute, Wayne State University, Detroit, MI
| |
Collapse
|
33
|
Chen M, Zhao J, Ali IHA, Marry S, Augustine J, Bhuckory M, Lynch A, Kissenpfennig A, Xu H. Cytokine Signaling Protein 3 Deficiency in Myeloid Cells Promotes Retinal Degeneration and Angiogenesis through Arginase-1 Up-Regulation in Experimental Autoimmune Uveoretinitis. THE AMERICAN JOURNAL OF PATHOLOGY 2018; 188:1007-1020. [PMID: 29452101 DOI: 10.1016/j.ajpath.2017.12.021] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/02/2017] [Revised: 11/14/2017] [Accepted: 12/07/2017] [Indexed: 01/01/2023]
Abstract
The suppressor of cytokine signaling protein 3 (SOCS3) critically controls immune cell activation, although its role in macrophage polarization and function remains controversial. Using experimental autoimmune uveoretinitis (EAU) as a model, we show that inflammation-mediated retinal degeneration is exaggerated and retinal angiogenesis is accelerated in mice with SOCS3 deficiency in myeloid cells (LysMCre/+SOCS3fl/fl). At the acute stage of EAU, the population of infiltrating neutrophils was increased and the population of macrophages decreased in LysMCre/+SOCS3fl/fl mice compared with that in wild-type (WT) mice. Real-time RT-PCR showed that the expression of tumor necrosis factor-α, IL-1β, interferon-γ, granulocyte-macrophage colony-stimulating factor, and arginase-1 was significantly higher in the LysMCre/+SOCS3fl/fl EAU retina in contrast to the WT EAU retina. The percentage of arginase-1+ infiltrating cells was significantly higher in the LysMCre/+SOCS3fl/fl EAU retina than that in the WT EAU retina. In addition, bone marrow-derived macrophages and neutrophils from the LysMCre/+SOCS3fl/fl mice express significantly higher levels of chemokine (C-C motif) ligand 2 and arginase-1 compared with those from WT mice. Inhibition of arginase using an l-arginine analog amino-2-borono-6-hexanoic suppressed inflammation-induced retinal angiogenesis without affecting the severity of inflammation. Our results suggest that SOCS3 critically controls the phenotype and function of macrophages and neutrophils under inflammatory conditions and loss of SOCS3 promotes the angiogenic phenotype of the cells through up-regulation of arginase-1.
Collapse
Affiliation(s)
- Mei Chen
- Centre for Experimental Medicine, School of Medicine, Dentistry & Biomedical Science, Queen's University Belfast, Belfast, United Kingdom
| | - Jiawu Zhao
- Centre for Experimental Medicine, School of Medicine, Dentistry & Biomedical Science, Queen's University Belfast, Belfast, United Kingdom
| | - Imran H A Ali
- Centre for Experimental Medicine, School of Medicine, Dentistry & Biomedical Science, Queen's University Belfast, Belfast, United Kingdom
| | - Stephen Marry
- Centre for Experimental Medicine, School of Medicine, Dentistry & Biomedical Science, Queen's University Belfast, Belfast, United Kingdom
| | - Josy Augustine
- Centre for Experimental Medicine, School of Medicine, Dentistry & Biomedical Science, Queen's University Belfast, Belfast, United Kingdom
| | - Mohajeet Bhuckory
- Centre for Experimental Medicine, School of Medicine, Dentistry & Biomedical Science, Queen's University Belfast, Belfast, United Kingdom
| | - Aisling Lynch
- Centre for Experimental Medicine, School of Medicine, Dentistry & Biomedical Science, Queen's University Belfast, Belfast, United Kingdom
| | - Adrien Kissenpfennig
- Centre for Experimental Medicine, School of Medicine, Dentistry & Biomedical Science, Queen's University Belfast, Belfast, United Kingdom
| | - Heping Xu
- Centre for Experimental Medicine, School of Medicine, Dentistry & Biomedical Science, Queen's University Belfast, Belfast, United Kingdom.
| |
Collapse
|
34
|
Pearsall EA, Cheng R, Zhou K, Takahashi Y, Matlock HG, Vadvalkar SS, Shin Y, Fredrick TW, Gantner ML, Meng S, Fu Z, Gong Y, Kinter M, Humphries KM, Szweda LI, Smith LEH, Ma JX. PPARα is essential for retinal lipid metabolism and neuronal survival. BMC Biol 2017; 15:113. [PMID: 29183319 PMCID: PMC5706156 DOI: 10.1186/s12915-017-0451-x] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2017] [Accepted: 11/06/2017] [Indexed: 11/29/2022] Open
Abstract
Background Peroxisome proliferator activated receptor-alpha (PPARα) is a ubiquitously expressed nuclear receptor. The role of endogenous PPARα in retinal neuronal homeostasis is unknown. Retinal photoreceptors are the highest energy-consuming cells in the body, requiring abundant energy substrates. PPARα is a known regulator of lipid metabolism, and we hypothesized that it may regulate lipid use for oxidative phosphorylation in energetically demanding retinal neurons. Results We found that endogenous PPARα is essential for the maintenance and survival of retinal neurons, with Pparα-/- mice developing retinal degeneration first detected at 8 weeks of age. Using extracellular flux analysis, we identified that PPARα mediates retinal utilization of lipids as an energy substrate, and that ablation of PPARα ultimately results in retinal bioenergetic deficiency and neurodegeneration. This may be due to PPARα regulation of lipid transporters, which facilitate the internalization of fatty acids into cell membranes and mitochondria for oxidation and ATP production. Conclusion We identify an endogenous role for PPARα in retinal neuronal survival and lipid metabolism, and furthermore underscore the importance of fatty acid oxidation in photoreceptor survival. We also suggest PPARα as a putative therapeutic target for age-related macular degeneration, which may be due in part to decreased mitochondrial efficiency and subsequent energetic deficits. Electronic supplementary material The online version of this article (doi:10.1186/s12915-017-0451-x) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Elizabeth A Pearsall
- Department of Ophthalmology, Boston Children's Hospital, Harvard Medical School, Boston, MA, 02115, USA.,Department of Physiology, University of Oklahoma Health Sciences Center, 941 Stanton L. Young Blvd., BSEB 328B, Oklahoma City, OK, 73104, USA
| | - Rui Cheng
- Department of Physiology, University of Oklahoma Health Sciences Center, 941 Stanton L. Young Blvd., BSEB 328B, Oklahoma City, OK, 73104, USA
| | - Kelu Zhou
- Department of Physiology, University of Oklahoma Health Sciences Center, 941 Stanton L. Young Blvd., BSEB 328B, Oklahoma City, OK, 73104, USA
| | - Yusuke Takahashi
- Department of Physiology, University of Oklahoma Health Sciences Center, 941 Stanton L. Young Blvd., BSEB 328B, Oklahoma City, OK, 73104, USA.,Section of Diabetes and Endocrinology, Department of Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA
| | - H Greg Matlock
- Department of Physiology, University of Oklahoma Health Sciences Center, 941 Stanton L. Young Blvd., BSEB 328B, Oklahoma City, OK, 73104, USA
| | - Shraddha S Vadvalkar
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, 73104, USA
| | - Younghwa Shin
- Department of Physiology, University of Oklahoma Health Sciences Center, 941 Stanton L. Young Blvd., BSEB 328B, Oklahoma City, OK, 73104, USA
| | - Thomas W Fredrick
- Department of Ophthalmology, Boston Children's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Marin L Gantner
- The Lowy Medical Research Institute, La Jolla, CA, 92037, USA
| | - Steven Meng
- Department of Ophthalmology, Boston Children's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Zhongjie Fu
- Department of Ophthalmology, Boston Children's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Yan Gong
- Department of Ophthalmology, Boston Children's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Michael Kinter
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, 73104, USA
| | - Kenneth M Humphries
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, 73104, USA
| | - Luke I Szweda
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, 73104, USA
| | - Lois E H Smith
- Department of Ophthalmology, Boston Children's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Jian-Xing Ma
- Department of Physiology, University of Oklahoma Health Sciences Center, 941 Stanton L. Young Blvd., BSEB 328B, Oklahoma City, OK, 73104, USA. .,Section of Diabetes and Endocrinology, Department of Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA.
| |
Collapse
|
35
|
Dang LTH, Aburatani T, Marsh GA, Johnson BG, Alimperti S, Yoon CJ, Huang A, Szak S, Nakagawa N, Gomez I, Ren S, Read SK, Sparages C, Aplin AC, Nicosia RF, Chen C, Ligresti G, Duffield JS. Hyperactive FOXO1 results in lack of tip stalk identity and deficient microvascular regeneration during kidney injury. Biomaterials 2017; 141:314-329. [PMID: 28711779 DOI: 10.1016/j.biomaterials.2017.07.010] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2017] [Revised: 06/07/2017] [Accepted: 07/06/2017] [Indexed: 02/06/2023]
Abstract
Loss of the microvascular (MV) network results in tissue ischemia, loss of tissue function, and is a hallmark of chronic diseases. The incorporation of a functional vascular network with that of the host remains a challenge to utilizing engineered tissues in clinically relevant therapies. We showed that vascular-bed-specific endothelial cells (ECs) exhibit differing angiogenic capacities, with kidney microvascular endothelial cells (MVECs) being the most deficient, and sought to explore the underlying mechanism. Constitutive activation of the phosphatase PTEN in kidney MVECs resulted in impaired PI3K/AKT activity in response to vascular endothelial growth factor (VEGF). Suppression of PTEN in vivo resulted in microvascular regeneration, but was insufficient to improve tissue function. Promoter analysis of the differentially regulated genes in KMVECs suggests that the transcription factor FOXO1 is highly active and RNAseq analysis revealed that hyperactive FOXO1 inhibits VEGF-Notch-dependent tip-cell formation by direct and indirect inhibition of DLL4 expression in response to VEGF. Inhibition of FOXO1 enhanced angiogenesis in human bio-engineered capillaries, and resulted in microvascular regeneration and improved function in mouse models of injury-repair.
Collapse
Affiliation(s)
- Lan T H Dang
- Research & Development, Biogen, Cambridge, MA, USA.
| | - Takahide Aburatani
- Division of Nephrology, Departments of Medicine & Pathology, University of Washington, Seattle, USA; Institute of Stem Cell & Regenerative Medicine, University of Washington, Seattle, WA, USA
| | | | | | | | | | - Angela Huang
- Research & Development, Biogen, Cambridge, MA, USA
| | - Suzanne Szak
- Research & Development, Biogen, Cambridge, MA, USA
| | - Naoki Nakagawa
- Division of Nephrology, Departments of Medicine & Pathology, University of Washington, Seattle, USA; Institute of Stem Cell & Regenerative Medicine, University of Washington, Seattle, WA, USA
| | - Ivan Gomez
- Research & Development, Biogen, Cambridge, MA, USA
| | - Shuyu Ren
- Research & Development, Biogen, Cambridge, MA, USA
| | - Sarah K Read
- Research & Development, Biogen, Cambridge, MA, USA
| | | | - Alfred C Aplin
- Department of Pathology, University of Washington, Seattle, WA, USA
| | - Roberto F Nicosia
- Department of Pathology, University of Washington, Seattle, WA, USA; Pathology and Laboratory Medicine Service, VA Puget Sound Health Care System, Seattle, WA, USA
| | - Chris Chen
- Department of Bioengineering, Boston University, Boston, USA
| | | | - Jeremy S Duffield
- Research & Development, Biogen, Cambridge, MA, USA; Division of Nephrology, Departments of Medicine & Pathology, University of Washington, Seattle, USA; Institute of Stem Cell & Regenerative Medicine, University of Washington, Seattle, WA, USA.
| |
Collapse
|
36
|
Sun Y, Liu CH, Wang Z, Meng SS, Burnim SB, SanGiovanni JP, Kamenecka TM, Solt LA, Chen J. RORα modulates semaphorin 3E transcription and neurovascular interaction in pathological retinal angiogenesis. FASEB J 2017. [PMID: 28646017 DOI: 10.1096/fj.201700172r] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Pathological proliferation of retinal blood vessels commonly causes vision impairment in proliferative retinopathies, including retinopathy of prematurity. Dysregulated crosstalk between the vasculature and retinal neurons is increasingly recognized as a major factor contributing to the pathogenesis of vascular diseases. Class 3 semaphorins (SEMA3s), a group of neuron-secreted axonal and vascular guidance factors, suppress pathological vascular growth in retinopathy. However, the upstream transcriptional regulators that mediate the function of SEMA3s in vascular growth are poorly understood. Here we showed that retinoic acid receptor-related orphan receptor α (RORα), a nuclear receptor and transcription factor, is a novel transcriptional regulator of SEMA3E-mediated neurovascular coupling in a mouse model of oxygen-induced proliferative retinopathy. We found that genetic deficiency of RORα substantially induced Sema3e expression in retinopathy. Both RORα and SEMA3E were expressed in retinal ganglion cells. RORα directly bound to a specific ROR response element on the promoter of Sema3e and negatively regulated Sema3e promoter-driven luciferase expression. Suppression of Sema3e using adeno-associated virus 2 carrying short hairpin RNA targeting Sema3e promoted disoriented pathological neovascularization and partially abolished the inhibitory vascular effects of RORα deficiency in retinopathy. Our findings suggest that RORα is a novel transcriptional regulator of SEMA3E-mediated neurovascular coupling in pathological retinal angiogenesis.-Sun, Y., Liu, C.-H., Wang, Z., Meng, S. S., Burnim, S. B., SanGiovanni, J. P., Kamenecka, T. M., Solt, L. A., Chen, J. RORα modulates semaphorin 3E transcription and neurovascular interaction in pathological retinal angiogenesis.
Collapse
Affiliation(s)
- Ye Sun
- Department of Ophthalmology, Harvard Medical School, Boston Children's Hospital, Boston, Massachusetts, USA
| | - Chi-Hsiu Liu
- Department of Ophthalmology, Harvard Medical School, Boston Children's Hospital, Boston, Massachusetts, USA
| | - Zhongxiao Wang
- Department of Ophthalmology, Harvard Medical School, Boston Children's Hospital, Boston, Massachusetts, USA
| | - Steven S Meng
- Department of Ophthalmology, Harvard Medical School, Boston Children's Hospital, Boston, Massachusetts, USA
| | - Samuel B Burnim
- Department of Ophthalmology, Harvard Medical School, Boston Children's Hospital, Boston, Massachusetts, USA
| | - John Paul SanGiovanni
- Section of Nutritional Neuroscience, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, Maryland, USA.,Department of Biochemistry and Molecular and Cellular Biology, Georgetown School of Medicine, Washington, D.C., USA
| | - Theodore M Kamenecka
- Department of Molecular Medicine, The Scripps Research Institute, Jupiter, Florida, USA
| | - Laura A Solt
- Department of Immunology and Microbiology, The Scripps Research Institute, Jupiter, Florida, USA
| | - Jing Chen
- Department of Ophthalmology, Harvard Medical School, Boston Children's Hospital, Boston, Massachusetts, USA;
| |
Collapse
|
37
|
Bucher F, Walz JM, Bühler A, Aguilar E, Lange C, Diaz-Aguilar S, Martin G, Schlunck G, Agostini H, Friedlander M, Stahl A. CNTF Attenuates Vasoproliferative Changes Through Upregulation of SOCS3 in a Mouse-Model of Oxygen-Induced Retinopathy. Invest Ophthalmol Vis Sci 2017; 57:4017-26. [PMID: 27494343 PMCID: PMC4986766 DOI: 10.1167/iovs.15-18508] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
Purpose Retinal vascular disease represents a major cause for vision loss in the Western world. Recent research has shown that neuronal and vascular damage are closely related in retinal disease. Ciliary neurotrophic factor (CNTF) is a well-studied neurotrophic factor that is currently being tested in clinical trials for the treatment of retinal degenerative diseases and macular telangiectasia. However, little is known about its effect on retinal vasculature. In this study, we investigate the effects of CNTF in retinal neovascular disease using the mouse model of oxygen-induced retinopathy (OIR). Methods Newborn pups were exposed to 75% oxygen from postnatal day (P)7 to P12 and subsequently returned to room air. Ciliary neurotrophic factor was injected intravitreally at OIR P12 and the vaso-obliterated and neovascular areas were quantified at OIR P17. Immunohistochemistry, RNA, and protein analysis were used to identify CNTF-responsive cells. In vitro experiments were performed to analyze the effect of CNTF on endothelial and astroglial cells. Results In the OIR model, CNTF facilitated capillary regrowth and attenuated preretinal neovascularization in a dose-dependent manner. The protective effect of CNTF was mediated via activation of the JAK/STAT3/SOCS3 signaling pathway. Immunohistochemical studies identified endothelial cells among others as CNTF-responsive cells in the retina. In vitro studies confirmed the anti-angiogenic effect of CNTF on endothelial cell sprouting. Conclusions This study provides evidence for a therapeutic potential of CNTF beyond degenerative retinal disease. Vasoproliferative retinopathies may benefit from a CNTF-dependent and SOCS3-mediated angiomodulatory effect.
Collapse
Affiliation(s)
- Felicitas Bucher
- Eye Center Medical Center, Faculty of Medicine, University of Freiburg, Germany 2Department of Cell and Molecular Biology, The Scripps Research Institute, La Jolla, California, United States
| | - Johanna M Walz
- Eye Center Medical Center, Faculty of Medicine, University of Freiburg, Germany 3Department of Pharmacology and Toxicology, University of Regensburg, Regensburg, Germany
| | - Anima Bühler
- Eye Center Medical Center, Faculty of Medicine, University of Freiburg, Germany
| | - Edith Aguilar
- Department of Cell and Molecular Biology, The Scripps Research Institute, La Jolla, California, United States
| | - Clemens Lange
- Eye Center Medical Center, Faculty of Medicine, University of Freiburg, Germany
| | - Sophia Diaz-Aguilar
- Department of Cell and Molecular Biology, The Scripps Research Institute, La Jolla, California, United States
| | - Gottfried Martin
- Eye Center Medical Center, Faculty of Medicine, University of Freiburg, Germany
| | - Günther Schlunck
- Eye Center Medical Center, Faculty of Medicine, University of Freiburg, Germany
| | - Hansjürgen Agostini
- Eye Center Medical Center, Faculty of Medicine, University of Freiburg, Germany
| | - Martin Friedlander
- Department of Cell and Molecular Biology, The Scripps Research Institute, La Jolla, California, United States
| | - Andreas Stahl
- Eye Center Medical Center, Faculty of Medicine, University of Freiburg, Germany
| |
Collapse
|
38
|
Sun Y, Lin Z, Liu CH, Gong Y, Liegl R, Fredrick TW, Meng SS, Burnim SB, Wang Z, Akula JD, Pu WT, Chen J, Smith LEH. Inflammatory signals from photoreceptor modulate pathological retinal angiogenesis via c-Fos. J Exp Med 2017; 214:1753-1767. [PMID: 28465464 PMCID: PMC5461000 DOI: 10.1084/jem.20161645] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2016] [Revised: 02/02/2017] [Accepted: 03/22/2017] [Indexed: 11/04/2022] Open
Abstract
Pathological neovessels growing into the normally avascular photoreceptors cause vision loss in many eye diseases, such as age-related macular degeneration and macular telangiectasia. Ocular neovascularization is strongly associated with inflammation, but the source of inflammatory signals and the mechanisms by which these signals regulate the disruption of avascular privilege in photoreceptors are unknown. In this study, we found that c-Fos, a master inflammatory regulator, was increased in photoreceptors in a model of pathological blood vessels invading photoreceptors: the very low-density lipoprotein receptor-deficient (Vldlr-/- ) mouse. Increased c-Fos induced inflammatory cytokines interleukin 6 (IL-6) and tumor necrosis factor (TNF), leading to activation of signal transducer and activator of transcription 3 (STAT3) and increased TNFα-induced protein 3 (TNFAIP3) in Vldlr-/- photoreceptors. IL-6 activated the STAT3/vascular endothelial growth factor A (VEGFA) pathway directly, and elevated TNFAIP3 suppressed SOCS3 (suppressor of cytokine signaling 3)-activated STAT3/VEGFA indirectly. Inhibition of c-Fos using photoreceptor-specific AAV (adeno-associated virus)-hRK (human rhodopsin kinase)-sh_c-fos or a chemical inhibitor substantially reduced the pathological neovascularization and rescued visual function in Vldlr-/- mice. These findings suggested that the photoreceptor c-Fos controls blood vessel growth into the normally avascular photoreceptor layer through the inflammatory signal-induced STAT3/VEGFA pathway.
Collapse
Affiliation(s)
- Ye Sun
- Department of Ophthalmology, Harvard Medical School, Boston Children's Hospital, Boston, MA 02115
| | - Zhiqiang Lin
- Department of Cardiology, Harvard Medical School, Boston Children's Hospital, Boston, MA 02115
| | - Chi-Hsiu Liu
- Department of Ophthalmology, Harvard Medical School, Boston Children's Hospital, Boston, MA 02115
| | - Yan Gong
- Department of Ophthalmology, Harvard Medical School, Boston Children's Hospital, Boston, MA 02115
| | - Raffael Liegl
- Department of Ophthalmology, Harvard Medical School, Boston Children's Hospital, Boston, MA 02115
| | - Thomas W Fredrick
- Department of Ophthalmology, Harvard Medical School, Boston Children's Hospital, Boston, MA 02115
| | - Steven S Meng
- Department of Ophthalmology, Harvard Medical School, Boston Children's Hospital, Boston, MA 02115
| | - Samuel B Burnim
- Department of Ophthalmology, Harvard Medical School, Boston Children's Hospital, Boston, MA 02115
| | - Zhongxiao Wang
- Department of Ophthalmology, Harvard Medical School, Boston Children's Hospital, Boston, MA 02115
| | - James D Akula
- Department of Ophthalmology, Harvard Medical School, Boston Children's Hospital, Boston, MA 02115
| | - William T Pu
- Department of Cardiology, Harvard Medical School, Boston Children's Hospital, Boston, MA 02115.,Harvard Stem Cell Institute, Harvard University, Cambridge, MA 02138
| | - Jing Chen
- Department of Ophthalmology, Harvard Medical School, Boston Children's Hospital, Boston, MA 02115
| | - Lois E H Smith
- Department of Ophthalmology, Harvard Medical School, Boston Children's Hospital, Boston, MA 02115
| |
Collapse
|
39
|
Chikuma S, Kanamori M, Mise-Omata S, Yoshimura A. Suppressors of cytokine signaling: Potential immune checkpoint molecules for cancer immunotherapy. Cancer Sci 2017; 108:574-580. [PMID: 28188673 PMCID: PMC5406529 DOI: 10.1111/cas.13194] [Citation(s) in RCA: 77] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2016] [Revised: 01/30/2017] [Accepted: 02/05/2017] [Indexed: 12/19/2022] Open
Abstract
Inhibition of immune checkpoint molecules, PD‐1 and CTLA4, has been shown to be a promising cancer treatment. PD‐1 and CTLA4 inhibit TCR and co‐stimulatory signals. The third T cell activation signal represents the signals from the cytokine receptors. The cytokine interferon‐γ (IFNγ) plays an important role in anti‐tumor immunity by activating cytotoxic T cells (CTLs). Most cytokines use the Janus kinase (JAK)/signal transducer and activator of transcription (STAT) pathway, and the suppressors of cytokine signaling (SOCS) family of proteins are major negative regulators of the JAK/STAT pathway. Among SOCS proteins, CIS, SOCS1, and SOCS3 proteins can be considered the third immunocheckpoint molecules since they regulate cytokine signals that control the polarization of CD4+ T cells and the maturation of CD8+ T cells. This review summarizes recent progress on CIS, SOCS1, and SOCS3 in terms of their anti‐tumor immunity and potential applications.
Collapse
Affiliation(s)
- Shunsuke Chikuma
- Department of Microbiology and Immunology, Keio University School of Medicine, Shinjuku-ku, Tokyo, Japan
| | - Mitsuhiro Kanamori
- Department of Microbiology and Immunology, Keio University School of Medicine, Shinjuku-ku, Tokyo, Japan
| | - Setsuko Mise-Omata
- Department of Microbiology and Immunology, Keio University School of Medicine, Shinjuku-ku, Tokyo, Japan
| | - Akihiko Yoshimura
- Department of Microbiology and Immunology, Keio University School of Medicine, Shinjuku-ku, Tokyo, Japan
| |
Collapse
|
40
|
Significant changes in endogenous retinal gene expression assessed 1 year after a single intraocular injection of AAV-CNTF or AAV-BDNF. MOLECULAR THERAPY-METHODS & CLINICAL DEVELOPMENT 2016; 3:16078. [PMID: 27933306 PMCID: PMC5142514 DOI: 10.1038/mtm.2016.78] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/14/2016] [Revised: 09/27/2016] [Accepted: 10/12/2016] [Indexed: 12/17/2022]
Abstract
Use of viral vectors to deliver therapeutic genes to the central nervous system holds promise for the treatment of neurodegenerative diseases and neurotrauma. Adeno-associated viral (AAV) vectors encoding brain-derived neurotrophic factor (BDNF) or ciliary derived neurotrophic factor (CNTF) promote the viability and regeneration of injured adult rat retinal ganglion cells. However, these growth-inducing transgenes are driven by a constitutively active promoter, thus we examined whether long-term AAV-mediated secretion of BDNF or CNTF affected endogenous retinal gene expression. One year after the intravitreal injection of AAV-green fluorescent protein (GFP), bi-cistronic AAV-BDNF-GFP or AAV-CNTF-GFP, mRNA was extracted and analyzed using custom 96 well polymerase chain reaction arrays. Of 93 test genes, 56% showed significantly altered expression in AAV-BDNF-GFP and/or AAV-CNTF-GFP retinas compared with AAV-GFP controls. Of these genes, 73% showed differential expression in AAV-BDNF versus AAV-CNTF injected eyes. To focus on retinal ganglion cell changes, quantitative polymerase chain reaction was undertaken on mRNA (16 genes) obtained from fixed retinal sections in which the ganglion cell layer was enriched. The sign and extent of fold changes in ganglion cell layer gene expression differed markedly from whole retinal samples. Sustained and global alteration in endogenous mRNA expression after gene therapy should be factored into any interpretation of experimental/clinical outcomes, particularly when introducing factors into the central nervous system that require secretion to evoke functionality.
Collapse
|
41
|
Pharmacologic Activation of Wnt Signaling by Lithium Normalizes Retinal Vasculature in a Murine Model of Familial Exudative Vitreoretinopathy. THE AMERICAN JOURNAL OF PATHOLOGY 2016; 186:2588-600. [PMID: 27524797 DOI: 10.1016/j.ajpath.2016.06.015] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/11/2016] [Revised: 06/16/2016] [Accepted: 06/21/2016] [Indexed: 12/12/2022]
Abstract
Familial exudative vitreoretinopathy (FEVR) is characterized by delayed retinal vascular development, which promotes hypoxia-induced pathologic vessels. In severe cases FEVR may lead to retinal detachment and visual impairment. Genetic studies linked FEVR with mutations in Wnt signaling ligand or receptors, including low-density lipoprotein receptor-related protein 5 (LRP5) gene. Here, we investigated ocular pathologies in a Lrp5 knockout (Lrp5(-/-)) mouse model of FEVR and explored whether treatment with a pharmacologic Wnt activator lithium could bypass the genetic defects, thereby protecting against eye pathologies. Lrp5(-/-) mice displayed significantly delayed retinal vascular development, absence of deep layer retinal vessels, leading to increased levels of vascular endothelial growth factor and subsequent pathologic glomeruloid vessels, as well as decreased inner retinal visual function. Lithium treatment in Lrp5(-/-) mice significantly restored the delayed development of retinal vasculature and the intralaminar capillary networks, suppressed formation of pathologic glomeruloid structures, and promoted hyaloid vessel regression. Moreover, lithium treatment partially rescued inner-retinal visual function and increased retinal thickness. These protective effects of lithium were largely mediated through restoration of canonical Wnt signaling in Lrp5(-/-) retina. Lithium treatment also substantially increased vascular tubular formation in LRP5-deficient endothelial cells. These findings suggest that pharmacologic activation of Wnt signaling may help treat ocular pathologies in FEVR and potentially other defective Wnt signaling-related diseases.
Collapse
|
42
|
MicroRNAs in Hyperglycemia Induced Endothelial Cell Dysfunction. Int J Mol Sci 2016; 17:518. [PMID: 27070575 PMCID: PMC4848974 DOI: 10.3390/ijms17040518] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2016] [Revised: 03/17/2016] [Accepted: 03/22/2016] [Indexed: 01/15/2023] Open
Abstract
Hyperglycemia is closely associated with prediabetes and Type 2 Diabetes Mellitus. Hyperglycemia increases the risk of vascular complications such as diabetic retinopathy, diabetic nephropathy, peripheral vascular disease and cerebro/cardiovascular diseases. Under hyperglycemic conditions, the endothelial cells become dysfunctional. In this study, we investigated the miRNA expression changes in human umbilical vein endothelial cells exposed to different glucose concentrations (5, 10, 25 and 40 mM glucose) and at various time intervals (6, 12, 24 and 48 h). miRNA microarray analyses showed that there is a correlation between hyperglycemia induced endothelial dysfunction and miRNA expression. In silico pathways analyses on the altered miRNA expression showed that the majority of the affected biological pathways appeared to be associated to endothelial cell dysfunction and apoptosis. We found the expression of ten miRNAs (miR-26a-5p, -26b-5p, 29b-3p, -29c-3p, -125b-1-3p, -130b-3p, -140-5p, -192-5p, -221-3p and -320a) to increase gradually with increasing concentration of glucose. These miRNAs were also found to be involved in endothelial dysfunction. At least seven of them, miR-29b-3p, -29c-3p, -125b-1-3p, -130b-3p, -221-3p, -320a and -192-5p, can be correlated to endothelial cell apoptosis.
Collapse
|
43
|
Smith LEH, Sun Y, VanHook AM. Science Signaling
Podcast for 22 September 2015: SOCS3 prevents pathological retinal neovascularization. Sci Signal 2015. [DOI: 10.1126/scisignal.aad3734] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
SOCS3 activity in neural and glial cells of the retina prevents pathological retinal neovascularization.
Collapse
Affiliation(s)
- Lois E. H. Smith
- Department of Ophthalmology, Harvard Medical School, Boston Children's Hospital, 300 Longwood Ave, Boston, MA 02115, USA
| | - Ye Sun
- Department of Ophthalmology, Harvard Medical School, Boston Children's Hospital, 300 Longwood Ave, Boston, MA 02115, USA
| | - Annalisa M. VanHook
- Web Editor, Science Signaling, American Association for the Advancement of Science, 1200 New York Avenue, NW, Washington, DC 20005, USA
| |
Collapse
|