1
|
Shelton DA, Papania JT, Getz TE, Sellers JT, Giradot PE, Chrenek MA, Grossniklaus HE, Boatright JH, Nickerson JM. Loss of Pigment Epithelium Derived Factor Sensitizes C57BL/6J Mice to Light-Induced Retinal Damage. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.12.04.626802. [PMID: 39679905 PMCID: PMC11643041 DOI: 10.1101/2024.12.04.626802] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/17/2024]
Abstract
Purpose Pigment epithelium-derived factor (PEDF) is a neurotrophic glycoprotein secreted by the retinal pigment epithelium (RPE) that supports retinal photoreceptor health. Deficits in PEDF are associated with increased inflammation and retinal degeneration in aging and diabetic retinopathy. We hypothesized that light-induced stress in C57BL/6J mice deficient in PEDF would lead to increased retinal neuronal and RPE defects, impaired expression of neurotrophic factor Insulin-like growth factor 1 (IGF-1), and overactivation of Galectin-3-mediated inflammatory signaling. Methods C57BL/6J mice expressing the RPE65 M450/M450 allele were crossed to PEDF KO/KO and wildtype (PEDF +/+) littermates. Mice were exposed to 50,000 lux light for 5 hours to initiate acute damage. Changes in visual function outcomes were tracked via electroretinogram (ERG), confocal scanning laser ophthalmoscopy(cSLO), and spectral domain optical coherence tomography (SD-OCT) on days 3, 5, and 7 post-light exposure. Gene and protein expression of Galectin-3 were measured by digital drop PCR (ddPCR) and western blot. To further investigate the role of galectin-3 on visual outcomes and PEDF expression after damage, we also used a small-molecule inhibitor to reduce its activity. Results Following light damage, PEDF KO/KO mice showed more severe retinal thinning, impaired visual function (reduced a-, b-, and c-wave amplitudes), and increased Galectin-3 expressing immune cell infiltration compared to PEDF +/+. PEDF KO/KO mice had suppressed damage-associated increases in IGF-1 expression. Additionally, baseline Galectin-3 mRNA and protein expression were reduced in PEDF KO/KO mice compared to PEDF +/+. However, after light damage, Galectin-3 expression decreases in PEDF +/+ mice but increases in PEDF KO/KO mice without reaching PEDF +/+ levels. Galectin-3 inhibition worsens retinal degeneration, reduces PEDF expression in PEDF +/+ mice, and mimics the effects seen in PEDF knockouts. Conclusions Loss of PEDF alone does not elicit functional defects in C57BL/6J mice. However, under light stress, PEDF deficiency significantly increases severe retinal degeneration, visual deficits, Galectin-3 expression, and suppression of IGF-1 than PEDF +/+. PEDF deficiency reduced baseline expression of Galectin-3, and pharmacological inhibition of Galectin-3 worsens outcomes and suppresses PEDF expression in PEDF +/+, suggesting a novel co-regulatory relationship between the two proteins in mitigating light-induced retinal damage.
Collapse
Affiliation(s)
- Debresha A. Shelton
- Department of Ophthalmology, Emory University, Atlanta, Georgia, United States
| | - Jack T. Papania
- Department of Ophthalmology, Emory University, Atlanta, Georgia, United States
| | - Tatiana E. Getz
- Department of Ophthalmology, Emory University, Atlanta, Georgia, United States
| | - Jana T. Sellers
- Department of Ophthalmology, Emory University, Atlanta, Georgia, United States
| | - Preston E. Giradot
- Department of Ophthalmology, Emory University, Atlanta, Georgia, United States
| | - Micah A. Chrenek
- Department of Ophthalmology, Emory University, Atlanta, Georgia, United States
| | | | - Jeffrey H. Boatright
- Department of Ophthalmology, Emory University, Atlanta, Georgia, United States
- Atlanta Veterans Administration Center for Visual and Neurocognitive Rehabilitation, Decatur, Georgia, United States
| | - John M. Nickerson
- Department of Ophthalmology, Emory University, Atlanta, Georgia, United States
| |
Collapse
|
2
|
Jiang W, Yang L, Liang S. Association between insulin-like growth factor-1 and ocular surface parameters in obese prepubertal boys. Eur J Pediatr 2024; 183:4807-4816. [PMID: 39235603 DOI: 10.1007/s00431-024-05748-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/19/2024] [Revised: 08/07/2024] [Accepted: 08/26/2024] [Indexed: 09/06/2024]
Abstract
The study aimed to investigate the correlation between insulin-like growth factor 1 (IGF-1) and ocular surface parameters in obese prepubertal boys. Thirty obese prepubertal boys and 30 age- and gender-matched healthy controls underwent physical measurements, laboratory tests, and ocular surface assessments. The obese group showed lower IGF-1 levels (P = 0.001), reduced Schirmer I tear test (SIT) (P <0.001), and higher meibomian gland scores (meiboscore) compared to controls (P = 0.015). Bivariate analysis revealed a positive association between IGF-1 and SIT (r = 0.677, P < 0.001), and a negative association with between IGF-1 and meiboscore (r = - 0.487, P < 0.001). Multiple regression analysis indicated that IGF-1 (P < 0.001) and triglycerides (P = 0.028) independently influenced SIT. Logistic analysis showed a significant association between decreased IGF-1 and higher meiboscore values (OR 0.994, 95% confidence interval 0.988-1.000; P = 0.033). CONCLUSION The findings suggest that reduced IGF-1 in obese prepubertal boys is independently linked to decreased SIT and increased meiboscore, irrespective of obesity and traditional cardiovascular risk factors. This implies that monitoring ocular surface parameters in obese children might provide a new perspective for clinical practice to focus on. WHAT IS KNOWN • Obese children exhibit decreased levels of IGF-1, and this reduction in IGF-1 is associated with cardiovascular metabolic complications related to obesity. • Ocular surface tissues might act as targets for hormones, might experience local effects of these hormone. WHAT IS NEW • In prepubertal obese boys, the decrease in IGF-1 is independently linked to decreased SIT and increased meiboscore, irrespective of obesity and traditional cardiovascular risk factors. • This finding implies that monitoring ocular surface parameters in obese children might provide a new perspective for clinical practice to focus on.
Collapse
Affiliation(s)
- Wen Jiang
- Institute of Medical Sciences, The Second Hospital of Shandong University, Jinan, Shandong, China
| | - Lixia Yang
- Department of Ophthalmology, The Second Hospital of Shandong University, Jinan, Shandong, China.
| | - Shuang Liang
- Department of Pediatrics, The Second Hospital of Shandong University, 247 Beiyuan Main Street, Jinan, 250021, Shandong, China.
| |
Collapse
|
3
|
Rajala RVS, Rajala A. From Insight to Eyesight: Unveiling the Secrets of the Insulin-Like Growth Factor Axis in Retinal Health. Aging Dis 2024; 15:1994-2002. [PMID: 38300646 PMCID: PMC11346401 DOI: 10.14336/ad.2024.0128] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2024] [Accepted: 01/29/2024] [Indexed: 02/02/2024] Open
Abstract
Insulin-like growth factor-1 (IGF-1) plays a diverse role in the retina, exerting its effects in both normal and diseased conditions. Deficiency of IGF-1 in humans leads to issues such as microcephaly, mental retardation, deafness, and postnatal growth failure. IGF-1 is produced in the retinal pigment epithelium (RPE) and activates the IGF-1 receptor (IGF-1R) in photoreceptor cells. When IGF-1R is absent in rod cells, it results in the degeneration of photoreceptors, emphasizing the neuroprotective function of IGF signaling in these cells. Contrastingly, in neovascular age-related macular degeneration (AMD), there is an overexpression of both IGF-1 and IGF-1R in RPE. The mechanisms behind this altered regulation of IGF-1 in diseased states are currently unknown. This comprehensive review provides recent insights into the role of IGF-1 in the health and disease of the retina, raising several unanswered questions that still need further investigation.
Collapse
Affiliation(s)
- Raju V S Rajala
- Departments of Ophthalmology, University of Oklahoma Health Sciences Center, Oklahoma, USA
- Physiology, University of Oklahoma Health Sciences Center, Oklahoma, USA
- Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma, USA
- Dean McGee Eye Institute, Oklahoma, Oklahoma, USA
| | - Ammaji Rajala
- Departments of Ophthalmology, University of Oklahoma Health Sciences Center, Oklahoma, USA
- Dean McGee Eye Institute, Oklahoma, Oklahoma, USA
| |
Collapse
|
4
|
Hou L, Ma J, Feng X, Chen J, Dong BH, Xiao L, Zhang X, Guo B. Caffeic acid and diabetic neuropathy: Investigating protective effects and insulin-like growth factor 1 (IGF-1)-related antioxidative and anti-inflammatory mechanisms in mice. Heliyon 2024; 10:e32623. [PMID: 38975173 PMCID: PMC11225750 DOI: 10.1016/j.heliyon.2024.e32623] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Revised: 05/15/2024] [Accepted: 06/06/2024] [Indexed: 07/09/2024] Open
Abstract
Diabetic neuropathy (DN) represents a common and debilitating complication of diabetes, affecting a significant proportion of patients. Despite available treatments focusing on symptom management, there remains an unmet need for therapies that address the underlying pathophysiology. In pursuit of novel interventions, this study evaluated the therapeutic effects of caffeic acid-a natural phenolic compound prevalent in various foods-on diabetic neuropathy using a mouse model, particularly examining its interaction with the Insulin-like Growth Factor 1 (IGF-1) signaling pathway. Caffeic acid was administered orally at two dosages (5 mg/kg and 10 mg/kg), and a comprehensive set of outcomes including fasting blood glucose levels, body weight, sensory behavior, spinal cord oxidative stress markers, inflammatory cytokines, and components of the IGF-1 signaling cascade were assessed. Additionally, to determine the specific contribution of IGF-1 signaling to the observed benefits, IGF1R inhibitor Picropodophyllin (PPP) was co-administered with caffeic acid. Our results demonstrated that caffeic acid, at both dosages, effectively reduced hyperglycemia and alleviated sensory behavioral deficits in diabetic mice. This was accompanied by a marked decrease in oxidative stress markers and an increase in antioxidant enzyme activities within the spinal cord. Significantly lowered microglial activation and inflammatory cytokine expression highlighted the potent antioxidative and anti-inflammatory effects of caffeic acid. Moreover, increases in both serum and spinal levels of IGF-1, along with elevated phosphorylated IGF1R, implicated the IGF-1 signaling pathway as a mediator of caffeic acid's neuroprotective actions. The partial reversal of caffeic acid's benefits by PPP substantiated the pivotal engagement of IGF-1 signaling in mediating its effects. Our findings delineate the capability of caffeic acid to mitigate DN symptoms, particularly through reducing spinal oxidative stress and inflammation, and pinpoint the integral role of IGF-1 signaling in these protective mechanisms. The insights gleaned from this study not only position caffeic acid as a promising dietary adjunct for managing diabetic neuropathy but also highlight the therapeutic potential of targeting spinal IGF-1 signaling as part of a strategic treatment approach.
Collapse
Affiliation(s)
- Leina Hou
- Department of Anesthesiology, Shaanxi Provincial Cancer Hospital, Xi'an, 710049, China
| | - Jiaqi Ma
- Department of Radiology, Shaanxi Provincial Cancer Hospital, Xi'an, 710049, China
| | - Xugang Feng
- Department of General Surgery, Shaanxi Provincial Cancer Hospital, Xi'an, 710049, China
| | - Jing Chen
- Department of Medical Oncology, Shaanxi Provincial Cancer Hospital, Xi'an, 710049, China
| | - Bu-huai Dong
- Department of Anesthesiology, Xi'an Honghui Hospital, Xi'an Jiaotong University Health Science Center, Xi'an, 710049, China
| | - Li Xiao
- Department of Anesthesiology, Xi'an Honghui Hospital, Xi'an Jiaotong University Health Science Center, Xi'an, 710049, China
| | - Xi Zhang
- Department of Pediatric Neurology, Northwest Women and Children's Hospital, Xi'an, 710049, China
| | - Bin Guo
- Department of Anesthesiology, Xi'an Honghui Hospital, Xi'an Jiaotong University Health Science Center, Xi'an, 710049, China
| |
Collapse
|
5
|
Chen F, Lu K, Bai N, Hao Y, Wang H, Zhao X, Yue F. Oral administration of ellagic acid mitigates perioperative neurocognitive disorders, hippocampal oxidative stress, and neuroinflammation in aged mice by restoring IGF-1 signaling. Sci Rep 2024; 14:2509. [PMID: 38291199 PMCID: PMC10827749 DOI: 10.1038/s41598-024-53127-8] [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: 07/20/2023] [Accepted: 01/29/2024] [Indexed: 02/01/2024] Open
Abstract
This study investigates the potential of ellagic acid (EA), a phytochemical with antioxidant and anti-inflammatory properties, in managing perioperative neurocognitive disorders (PND). PND, which represents a spectrum of cognitive impairments often faced by elderly patients, is principally linked to surgical and anesthesia procedures, and heavily impacted by oxidative stress in the hippocampus and microglia-induced neuroinflammation. Employing an aged mice model subjected to abdominal surgery, we delve into EA's ability to counteract postoperative oxidative stress and cerebral inflammation by engaging the Insulin-like growth factor-1 (IGF-1) pathway. Our findings revealed that administering EA orally notably alleviated post-surgical cognitive decline in older mice, a fact that was manifested in improved performance during maze tests. This enhancement in the behavioral performance of the EA-treated mice corresponded with the rejuvenation of IGF-1 signaling, a decrease in oxidative stress markers in the hippocampus (like MDA and carbonylated protein), and an increase in the activity of antioxidant enzymes such as SOD and CAT. Alongside these, we observed a decrease in microglia-driven neuroinflammation in the hippocampus, thus underscoring the antioxidant and anti-inflammatory roles of EA. Interestingly, when EA was given in conjunction with an IGF1R inhibitor, these benefits were annulled, accentuating the pivotal role that the IGF-1 pathway plays in the neuroprotective potential of EA. Hence, EA could serve as a potent candidate for safeguarding against PND in older patients by curbing oxidative stress and neuroinflammation through the activation of the IGF-1 pathway.
Collapse
Affiliation(s)
- Fang Chen
- Department of Anesthesiology, Shaanxi Provincial People's Hospital, The Third Affiliated Hospital of Xi'an Jiaotong University, Xi'an Jiaotong University, Xi'an, 710068, Shaanxi, China
| | - Kai Lu
- Department of Anesthesiology, Shaanxi Provincial People's Hospital, The Third Affiliated Hospital of Xi'an Jiaotong University, Xi'an Jiaotong University, Xi'an, 710068, Shaanxi, China
| | - Ning Bai
- Department of Anesthesiology, Shaanxi Provincial People's Hospital, The Third Affiliated Hospital of Xi'an Jiaotong University, Xi'an Jiaotong University, Xi'an, 710068, Shaanxi, China
| | - Yabo Hao
- Department of Anesthesiology, Shaanxi Provincial People's Hospital, The Third Affiliated Hospital of Xi'an Jiaotong University, Xi'an Jiaotong University, Xi'an, 710068, Shaanxi, China
| | - Hui Wang
- Department of Anesthesiology, Shaanxi Provincial People's Hospital, The Third Affiliated Hospital of Xi'an Jiaotong University, Xi'an Jiaotong University, Xi'an, 710068, Shaanxi, China
| | - Xinrong Zhao
- Department of Anesthesiology, Shaanxi Provincial People's Hospital, The Third Affiliated Hospital of Xi'an Jiaotong University, Xi'an Jiaotong University, Xi'an, 710068, Shaanxi, China
| | - Fang Yue
- Department of Anesthesiology, Shaanxi Provincial People's Hospital, The Third Affiliated Hospital of Xi'an Jiaotong University, Xi'an Jiaotong University, Xi'an, 710068, Shaanxi, China.
| |
Collapse
|
6
|
Shigesada N, Shikada N, Shirai M, Toriyama M, Higashijima F, Kimura K, Kondo T, Bessho Y, Shinozuka T, Sasai N. Combination of blockade of endothelin signalling and compensation of IGF1 expression protects the retina from degeneration. Cell Mol Life Sci 2024; 81:51. [PMID: 38252153 PMCID: PMC10803390 DOI: 10.1007/s00018-023-05087-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2023] [Revised: 12/01/2023] [Accepted: 12/12/2023] [Indexed: 01/23/2024]
Abstract
Retinitis pigmentosa (RP) and macular dystrophy (MD) cause severe retinal dysfunction, affecting 1 in 4000 people worldwide. This disease is currently assumed to be intractable, because effective therapeutic methods have not been established, regardless of genetic or sporadic traits. Here, we examined a RP mouse model in which the Prominin-1 (Prom1) gene was deficient and investigated the molecular events occurring at the outset of retinal dysfunction. We extracted the Prom1-deficient retina subjected to light exposure for a short time, conducted single-cell expression profiling, and compared the gene expression with and without stimuli. We identified the cells and genes whose expression levels change directly in response to light stimuli. Among the genes altered by light stimulation, Igf1 was decreased in rod photoreceptor cells and astrocytes under the light-stimulated condition. Consistently, the insulin-like growth factor (IGF) signal was weakened in light-stimulated photoreceptor cells. The recovery of Igf1 expression with the adeno-associated virus (AAV) prevented photoreceptor cell death, and its treatment in combination with the endothelin receptor antagonist led to the blockade of abnormal glial activation and the promotion of glycolysis, thereby resulting in the improvement of retinal functions, as assayed by electroretinography. We additionally demonstrated that the attenuation of mammalian/mechanistic target of rapamycin (mTOR), which mediates IGF signalling, leads to complications in maintaining retinal homeostasis. Together, we propose that combinatorial manipulation of distinct mechanisms is useful for the maintenance of the retinal condition.
Collapse
Affiliation(s)
- Naoya Shigesada
- Division of Biological Science, Nara Institute of Science and Technology, Ikoma, 630-0192, Japan
| | - Naoya Shikada
- Division of Biological Science, Nara Institute of Science and Technology, Ikoma, 630-0192, Japan
| | - Manabu Shirai
- Omics Research Center (ORC), National Cerebral and Cardiovascular Center, Suita, Osaka, 564-8565, Japan
| | - Michinori Toriyama
- Department of Biomedical Chemistry, School of Science and Technology, Kwansei Gakuin University, Sanda, 669-1337, Japan
| | - Fumiaki Higashijima
- Department of Ophthalmology, Graduate School of Medicine, Yamaguchi University, Ube, 755-0046, Japan
| | - Kazuhiro Kimura
- Department of Ophthalmology, Graduate School of Medicine, Yamaguchi University, Ube, 755-0046, Japan
| | - Toru Kondo
- Division of Stem Cell Biology, Institute for Genetic Medicine, Hokkaido University, Sapporo, 060-0815, Japan
| | - Yasumasa Bessho
- Division of Biological Science, Nara Institute of Science and Technology, Ikoma, 630-0192, Japan
| | - Takuma Shinozuka
- Division of Biological Science, Nara Institute of Science and Technology, Ikoma, 630-0192, Japan
| | - Noriaki Sasai
- Division of Biological Science, Nara Institute of Science and Technology, Ikoma, 630-0192, Japan.
| |
Collapse
|
7
|
Beltramo E, Mazzeo A, Porta M. Release of Pro-Inflammatory/Angiogenic Factors by Retinal Microvascular Cells Is Mediated by Extracellular Vesicles Derived from M1-Activated Microglia. Int J Mol Sci 2023; 25:15. [PMID: 38203187 PMCID: PMC10778795 DOI: 10.3390/ijms25010015] [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: 10/25/2023] [Revised: 12/11/2023] [Accepted: 12/17/2023] [Indexed: 01/12/2024] Open
Abstract
The interactions between the neuronal and vascular sides of the retina during diabetic retinopathy (DR) have gained increasing attention. Microglia is responsible for the immune response to inflammation inside the retina, which could be mediated by paracrine signals carried by extracellular vesicles (EVs). We aimed to characterize EVs released from immortalized human microglial cells in inflammation and investigate their effects on the retinal microvasculature and the anti-inflammatory potential of thiamine in this context. M1 pro-inflammatory polarization in microglia was induced through a cytokine cocktail. EVs were isolated from the supernatants, characterized, and used to stimulate human retinal endothelial cells (HRECs) and pericytes (HRPs). Microvascular cell functions and their release of pro-inflammatory/angiogenic factors were assessed. M1-derived EVs showed increased content of miR-21, miR-155, CCL2, MMP2, and MMP9, and enhanced apoptosis, proliferation, migration, and ROS production in HRPs and HRECs. IL-1β, IL-6, MMP9, CCL2, and VEGF release increased in HRPs exposed to M1-derived EVs, while HRECs showed augmented IL-6, Ang2, VEGF, and PDFG-B. Addition of thiamine to M1-microglial cultures reverted most of these effects. In conclusion, M1-derived EVs stimulate functional changes and secretion of pro-inflammatory/angiogenic molecules in microvascular cells, exacerbating inflammatory damage and retinopathy features. Thiamine added to microglia exerts anti-inflammatory effects.
Collapse
Affiliation(s)
- Elena Beltramo
- Department of Medical Sciences, University of Turin, 10126 Torino, Italy; (A.M.); (M.P.)
| | | | | |
Collapse
|
8
|
Puddu A, Maggi DC. Klotho: A new therapeutic target in diabetic retinopathy? World J Diabetes 2023; 14:1027-1036. [PMID: 37547589 PMCID: PMC10401458 DOI: 10.4239/wjd.v14.i7.1027] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/16/2023] [Revised: 05/12/2023] [Accepted: 05/22/2023] [Indexed: 07/12/2023] Open
Abstract
Klotho (Kl) is considered an antiaging gene, mainly for the inhibition of the insulin-like growth factor-1 signaling. Kl exists as full-length transmembrane, which acts as co-receptor for fibroblast growth factor receptor, and in soluble forms (sKl). The sKl may exert pleiotropic effects on organs and tissues by regulating several pathways involved in the pathogenesis of diseases associated with oxidative and inflammatory state. In diabetic Patients, serum levels of Kl are significantly decreased compared to healthy subjects, and are related to duration of diabetes. In diabetic retinopathy (DR), one of the most common microvascular complications of type 2 diabetes, serum Kl levels are negatively correlated with progression of the disease. A lot of evidences showed that Kl regulates several mechanisms involved in maintaining homeostasis and functions of retinal cells, including phagocytosis, calcium signaling, secretion of vascular endothelial growth factor A (VEGF-A), maintenance of redox status, and melanin biosynthesis. Experimental data have been shown that Kl exerts positive effects on several mechanisms involved in onset and progression of DR. In particular, treatment with Kl: (1) Prevents apoptosis induced by oxidative stress in human retinal endothelial cells and in retinal pigment epithelium (RPE) cells; (2) reduces secretion of VEGF-A by RPE cells; and (3) decreases subretinal fibrosis and preserves autophagic activity. Therefore, Kl may become a novel biomarker and a good candidate for the treatment of DR.
Collapse
Affiliation(s)
- Alessandra Puddu
- Department of Internal Medicine and Medical Specialties, University of Genova, Genova 16132, Italy
| | - Davide Carlo Maggi
- Department of Internal Medicine and Medical Specialties, University of Genova, Genova 16132, Italy
| |
Collapse
|
9
|
Du H, Xia J, Huang L, Zheng L, Gu W, Yi F. Relationship between insulin-like growth factor-1 and cerebral small vessel disease and its mechanisms: advances in the field. Front Aging Neurosci 2023; 15:1190869. [PMID: 37358957 PMCID: PMC10285072 DOI: 10.3389/fnagi.2023.1190869] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Accepted: 05/23/2023] [Indexed: 06/28/2023] Open
Abstract
Insulin-like growth factor-1 (IGF-1) is an active polypeptide protein that closely resembles the structural sequence of insulin and is involved in a variety of metabolic processes in the body. Decreased IGF-1 circulation levels are associated with an increased risk of stroke and a poorer prognosis, but the relationship with cerebral small vessel disease (cSVD) is unclear. Some studies found that the level of IGF-1 in patients with cSVD was significantly reduced, but the clinical significance and underlying mechanisms are unknown. This article reviews the correlation between IGF-1 and cerebrovascular disease and explores the potential relationship and mechanism between IGF-1 and cSVD.
Collapse
Affiliation(s)
- Hao Du
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Jian Xia
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Clinical Research Center for Cerebrovascular Disease of Hunan Province, Central South University, Changsha, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Lei Huang
- Department of Rehabilitation, Hunan Provincial People’s Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha, Hunan, China
| | - Lan Zheng
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Wenping Gu
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Clinical Research Center for Cerebrovascular Disease of Hunan Province, Central South University, Changsha, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Fang Yi
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Department of Geriatric Neurology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| |
Collapse
|
10
|
Shi Y, Liu Y, Wu C, Liu X, Hu W, Yang Z, Li Z, Li Y, Deng C, Wei K, Gu C, Chen X, Su W, Zhuo Y. N,N-Dimethyl-3β-hydroxycholenamide attenuates neuronal death and retinal inflammation in retinal ischemia/reperfusion injury by inhibiting Ninjurin 1. J Neuroinflammation 2023; 20:91. [PMID: 37029422 PMCID: PMC10082498 DOI: 10.1186/s12974-023-02754-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Accepted: 03/01/2023] [Indexed: 04/09/2023] Open
Abstract
BACKGROUND Retinal ischemia-reperfusion (RIR) injury refers to an obstruction in the retinal blood supply followed by reperfusion. Although the molecular mechanism underlying the ischemic pathological cascade is not fully understood, neuroinflammation plays a crucial part in the mortality of retinal ganglion cells. METHODS Single-cell RNA sequencing (scRNA-seq), molecular docking, and transfection assay were used to explore the effectiveness and pathogenesis of N,N-dimethyl-3β-hydroxycholenamide (DMHCA)-treated mice with RIR injury and DMHCA-treated microglia after oxygen and glucose deprivation/reoxygenation (OGD/R). RESULTS DMHCA could suppress inflammatory gene expression and attenuate neuronal lesions, restoring the retinal structure in vivo. Using scRNA-seq on the retina of DMHCA-treated mice, we provided novel insights into RIR immunity and demonstrated nerve injury-induced protein 1 (Ninjurin1/Ninj 1) as a promising treatment target for RIR. Moreover, the expression of Ninj1, which was increased in RIR injury and OGD/R-treated microglia, was downregulated in the DMHCA-treated group. DMHCA suppressed the activation of the nuclear factor kappa B (NF-κB) pathways induced by OGD/R, which was undermined by the NF-κB pathway agonist betulinic acid. Overexpressed Ninj1 reversed the anti-inflammatory and anti-apoptotic function of DMHCA. Molecular docking indicated that for Ninj1, DMHCA had a low binding energy of - 6.6 kcal/mol, suggesting highly stable binding. CONCLUSION Ninj1 may play a pivotal role in microglia-mediated inflammation, while DMHCA could be a potential treatment strategy against RIR injury.
Collapse
Affiliation(s)
- Yunhong Shi
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Sun Yat-Sen University, No. 7 Jinsui Road, Tianhe District, Guangzhou, 510060, Guangdong, China
| | - Yidan Liu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Sun Yat-Sen University, No. 7 Jinsui Road, Tianhe District, Guangzhou, 510060, Guangdong, China
| | - Caiqing Wu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Sun Yat-Sen University, No. 7 Jinsui Road, Tianhe District, Guangzhou, 510060, Guangdong, China
| | - Xiuxing Liu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Sun Yat-Sen University, No. 7 Jinsui Road, Tianhe District, Guangzhou, 510060, Guangdong, China
| | - Wenfei Hu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Sun Yat-Sen University, No. 7 Jinsui Road, Tianhe District, Guangzhou, 510060, Guangdong, China
| | - Zhenlan Yang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Sun Yat-Sen University, No. 7 Jinsui Road, Tianhe District, Guangzhou, 510060, Guangdong, China
| | - Zhidong Li
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Sun Yat-Sen University, No. 7 Jinsui Road, Tianhe District, Guangzhou, 510060, Guangdong, China
| | - Yangyang Li
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Sun Yat-Sen University, No. 7 Jinsui Road, Tianhe District, Guangzhou, 510060, Guangdong, China
| | - Caibin Deng
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Sun Yat-Sen University, No. 7 Jinsui Road, Tianhe District, Guangzhou, 510060, Guangdong, China
| | - Kun Wei
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Sun Yat-Sen University, No. 7 Jinsui Road, Tianhe District, Guangzhou, 510060, Guangdong, China
| | - Chenyang Gu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Sun Yat-Sen University, No. 7 Jinsui Road, Tianhe District, Guangzhou, 510060, Guangdong, China
| | - Xuhao Chen
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Sun Yat-Sen University, No. 7 Jinsui Road, Tianhe District, Guangzhou, 510060, Guangdong, China
| | - Wenru Su
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Sun Yat-Sen University, No. 7 Jinsui Road, Tianhe District, Guangzhou, 510060, Guangdong, China.
| | - Yehong Zhuo
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Sun Yat-Sen University, No. 7 Jinsui Road, Tianhe District, Guangzhou, 510060, Guangdong, China.
| |
Collapse
|
11
|
Huryn LA, Kozycki CT, Serpen JY, Zein WM, Ullah E, Iannaccone A, Williams LB, Sobrin L, Brooks BP, Sen HN, Hufnagel RB, Kastner DL, Kodati S. Ophthalmic Manifestations of ROSAH (Retinal Dystrophy, Optic Nerve Edema, Splenomegaly, Anhidrosis, and Headache) Syndrome, an Inherited NF κB-Mediated Autoinflammatory Disease with Retinal Dystrophy. Ophthalmology 2023; 130:423-432. [PMID: 36332842 PMCID: PMC10038920 DOI: 10.1016/j.ophtha.2022.10.026] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Revised: 10/07/2022] [Accepted: 10/26/2022] [Indexed: 11/06/2022] Open
Abstract
PURPOSE We aimed to characterize the ocular phenotype of patients with ROSAH (retinal dystrophy, optic nerve edema, splenomegaly, anhidrosis, and headache) syndrome and their response to therapy. DESIGN Single-center observational case study. PARTICIPANTS Eleven patients with a diagnosis of ROSAH syndrome and mutation in ALPK1 were included. METHODS Patients with molecularly confirmed ROSAH syndrome underwent ophthalmic evaluation, including visual acuity testing, slit-lamp and dilated examinations, color fundus and autofluorescence imaging, fluorescein angiography, OCT, and electrophysiologic testing. MAIN OUTCOME MEASURES Visual acuity, electrophysiology, fluorescein angiography, and OCT findings. RESULTS Eleven individuals (6 female and 5 male patients) from 7 families ranging in age from 7.3 to 60.2 years at the time of the initial evaluation were included in this study. Seven patients were followed up for a mean of 2.6 years (range, 0.33-5.0 years). Best-corrected visual acuity at baseline ranged from 20/16 to no light perception. Variable signs or sequelae of intraocular inflammation were observed in 9 patients, including keratic precipitates, band keratopathy, trace to 2+ anterior chamber cells, cystoid macular edema, and retinal vasculitis on fluorescein angiography. Ten patients were observed to show optic disc elevation and demonstrated peripapillary thickening on OCT. Seven patients showed retinal degeneration consistent with a cone-rod dystrophy, with atrophy tending to involve the posterior pole and extending peripherally. One patient with normal electroretinography findings and visual evoked potential was found to have decreased Arden ratio on electro-oculography. CONCLUSIONS Leveraging insights from the largest single-center ROSAH cohort described to date, this study identified 3 main factors as contributing to changes in visual function of patients with ROSAH syndrome: optic nerve involvement; intraocular inflammation, including cystoid macular edema; and retinal degeneration. More work is needed to determine how to arrest the progressive vision loss associated with ROSAH syndrome. FINANCIAL DISCLOSURE(S) Proprietary or commercial disclosure may be found after the references.
Collapse
Affiliation(s)
- Laryssa A Huryn
- National Eye Institute, National Institutes of Health, Bethesda, Maryland.
| | - Christina Torres Kozycki
- National Human Genome Research Institute, National Institutes of Health, Bethesda, Maryland; National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland
| | - Jasmine Y Serpen
- National Eye Institute, National Institutes of Health, Bethesda, Maryland
| | - Wadih M Zein
- National Eye Institute, National Institutes of Health, Bethesda, Maryland
| | - Ehsan Ullah
- National Eye Institute, National Institutes of Health, Bethesda, Maryland
| | | | - Lloyd B Williams
- Duke Eye Center, Duke University School of Medicine, Durham, North Carolina
| | - Lucia Sobrin
- Massachusetts Eye and Ear Infirmary, Boston, Massachusetts
| | - Brian P Brooks
- National Eye Institute, National Institutes of Health, Bethesda, Maryland
| | - H Nida Sen
- National Eye Institute, National Institutes of Health, Bethesda, Maryland
| | - Robert B Hufnagel
- National Eye Institute, National Institutes of Health, Bethesda, Maryland
| | - Daniel L Kastner
- National Human Genome Research Institute, National Institutes of Health, Bethesda, Maryland
| | - Shilpa Kodati
- National Eye Institute, National Institutes of Health, Bethesda, Maryland
| |
Collapse
|
12
|
Cellular and Molecular Mechanisms of Pathogenesis Underlying Inherited Retinal Dystrophies. Biomolecules 2023; 13:biom13020271. [PMID: 36830640 PMCID: PMC9953031 DOI: 10.3390/biom13020271] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Revised: 01/23/2023] [Accepted: 01/27/2023] [Indexed: 02/04/2023] Open
Abstract
Inherited retinal dystrophies (IRDs) are congenital retinal degenerative diseases that have various inheritance patterns, including dominant, recessive, X-linked, and mitochondrial. These diseases are most often the result of defects in rod and/or cone photoreceptor and retinal pigment epithelium function, development, or both. The genes associated with these diseases, when mutated, produce altered protein products that have downstream effects in pathways critical to vision, including phototransduction, the visual cycle, photoreceptor development, cellular respiration, and retinal homeostasis. The aim of this manuscript is to provide a comprehensive review of the underlying molecular mechanisms of pathogenesis of IRDs by delving into many of the genes associated with IRD development, their protein products, and the pathways interrupted by genetic mutation.
Collapse
|
13
|
Prokosch V, Li P, Shi X. Glaucoma as a Neurodegenerative and Inflammatory Disease. Klin Monbl Augenheilkd 2023; 240:125-129. [PMID: 36265500 DOI: 10.1055/a-1965-0044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Glaucoma is a neurodegenerative disease that leads to irreversible loss of vision through degeneration of the retinal ganglia cells (RGCs). Glaucoma is one of the most frequent causes of blindness in the world. Intraocular pressure is the main risk factor for the occurrence and development of this disease. Treatment is largely based on reducing internal optical pressure. However, some patients may deteriorate or become blind, despite normal or reduced internal optical pressure. The pathophysiological details are still unclear. Neuroinflammatory processes are also apparently an additional cause. In principle, innate or local responses of the adaptive immune system can be distinguished. The reaction of the innate immune system, particularly the local microglial cells, has long been studied. The macroglia with the astrocytes and Müller cells and their homeostatic effects have also long been known. On the other hand, it has long been thought that the retina with its RGZs was inert to adaptive immunological reactions - due to the function of the blood brain barrier. However, this system may be disturbed by antigen presentation, leading to a reaction of the adaptive immune system, with B cell and T cell responses. In this context, the key proteins are presumably heat shock proteins. We now know that neuroinflammation is important in glaucoma, as in other neurodegenerative diseases. It is important to increase our understanding of these phenomena. In this review article, we present our current knowledge of the role of the micro- and macroglia, the adaptive immune system, and the heat shock proteins.
Collapse
Affiliation(s)
- Verena Prokosch
- Department of Ophthalmology, Faculty of Medicine and University Hospital of Cologne, University of Cologne, Germany
| | - Panpan Li
- Department of Ophthalmology, Faculty of Medicine and University Hospital of Cologne, University of Cologne, Germany
| | - Xin Shi
- Department of Ophthalmology, Faculty of Medicine and University Hospital of Cologne, University of Cologne, Germany
| |
Collapse
|
14
|
Truong T, Silkiss RZ. The Role of Insulin-like Growth Factor-1 and Its Receptor in the Eye: A Review and Implications for IGF-1R Inhibition. Ophthalmic Plast Reconstr Surg 2023; 39:4-12. [PMID: 36598389 DOI: 10.1097/iop.0000000000002146] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
PURPOSE FDA approval of teprotumumab for thyroid eye disease in January 2020 reinforced interest in the pharmacologic potential of insulin-like growth factor-1 (IGF-1) and its receptor, IGF-1R. Despite recent approval and adaptation for ophthalmic use, IGF-1R inhibitors are not a new therapeutic class. In 1986, Yamashita described aIR3, a monoclonal antibody to IGF-1R (anti-IGF-1R), that inhibited the effect of IGF-1 on growth hormone release. Given the widespread presence of IGF-1R, interrupting this receptor can lead to systemic physiologic effects, some adverse. We aim to review what is known about IGF-1/IGF-1R in the eye and consider the possible local side effects, unintended consequences, and potential uses of this medication class. METHODS A PubMed database search utilizing the keywords "insulin-like growth factor-1, eye, inhibitor, antibody, side effect" was performed to identify publications discussing IGF-1 in the human eye from January 2011 to August 2021. Criteria for acceptance included studies discussing human subjects or human tissue specifically related to the eye. RESULTS Out of a total of 230 articles, 47 were organized in 3 subject groups for discussion: thyroid-associated orbitopathy, cornea and the ocular surface, and the retina and neovascularization. Review of the literature demonstrated that IGF-1 affects growth and development of the eye, epithelial proliferation, retinal angiogenesis, inflammation, and is associated with thyroid-associated orbitopathy. CONCLUSIONS IGF-1R exists throughout in the human body, including the cornea, retina, and orbit. Research regarding ocular effects of IGF-1/IGF-1R outside thyroid eye disease is limited. Carefully designed studies and clinical assessments of patients undergoing treatment with anti-IGF-1R may identify ocular side effects and foster consideration of the role of anti-IGF-1R in ocular therapeutics. Given the increasing use of anti-IGF-1R antibodies, understanding their ocular effects, side effects, and potential systemic implications for use in disease is critical.
Collapse
Affiliation(s)
- Timothy Truong
- Department of Ophthalmology, Division of Oculofacial Plastic Surgery, California Pacific Medical Center, San Francisco, California, U.S.A
| | - Rona Z Silkiss
- Department of Ophthalmology, Division of Oculofacial Plastic Surgery, California Pacific Medical Center, San Francisco, California, U.S.A
- Department of Ophthalmology, Division of Oculofacial Plastic Surgery, Silkiss Eye Surgery, San Francisco, California, U.S.A
| |
Collapse
|
15
|
Hamilton OS, Steptoe A. Socioeconomic determinants of inflammation and neuroendocrine activity: A longitudinal analysis of compositional and contextual effects. Brain Behav Immun 2023; 107:276-285. [PMID: 36270438 DOI: 10.1016/j.bbi.2022.10.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Revised: 10/08/2022] [Accepted: 10/13/2022] [Indexed: 11/06/2022] Open
Abstract
Socioeconomic determinants are well-established modulators of inflammation and neuroendocrine activity. Less clear is whether neighbourhood-contextual or individual-compositional factors are more closely associated with gradients in these biomarkers. Here, we examine how immune and neuroendocrine activity are cross-sectionally and longitudinally nested in meso-level socioeconomic characteristics. Participants, male and female, aged ≥50, were recruited from the English Longitudinal Study of Ageing (ELSA). Neighbourhood (Index of Multiple Deprivation [IMD]) and individual (Wealth/Education/Occupational Social Class [Occupation]) factors were drawn from wave 4 (baseline; 2008). Immune and neuroendocrine biomarkers (indexed by C-reactive protein [CRP; n = 3,968]; fibrinogen [n = 3,932]; white blood cell counts [WBCC; n = 4,022]; insulin-like growth factor-1 [IGF-1; n = 4,056]) were measured at baseline and 4-years later (wave 6; 2012). Covariates at baseline included demographic, clinical, and lifestyle variables. Lower socioeconomic status was associated with heighted inflammation and lower neuroendocrine activity unadjusted both cross-sectionally and longitudinally. With few exceptions, cross-sectional associations remained significant after full adjustment. Prospectively, low IMD remained associated with higher CRP and WBCC; wealth with WBCC; and education and occupation with fibrinogen and WBCC. IMD-biomarker associations were reduced when wealth was simultaneously taken into account. Lifestyle accounted for the greatest variance in associations between socioeconomic indicators and inflammation (≤42.11%), but demographics were more salient to neuroendocrine activity (≤88.46%). Neighbourhood-contextual factors were stronger indicators of aberrant biomarker activity than individual-compositional factors in cross-sectional analyses but were largely explained by wealth differences prospectively. Therefore, immune and neuroendocrine changes depended on the composition of the population living in an area, rather than the area itself.
Collapse
Affiliation(s)
- Odessa S Hamilton
- Department of Behavioural Science and Health, University College London, 1-19 Torrington Place, London WC1E 7HB, UK.
| | - Andrew Steptoe
- Department of Behavioural Science and Health, University College London, 1-19 Torrington Place, London WC1E 7HB, UK
| |
Collapse
|
16
|
Wei L, Sun X, Fan C, Li R, Zhou S, Yu H. The pathophysiological mechanisms underlying diabetic retinopathy. Front Cell Dev Biol 2022; 10:963615. [PMID: 36111346 PMCID: PMC9468825 DOI: 10.3389/fcell.2022.963615] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Accepted: 08/12/2022] [Indexed: 12/04/2022] Open
Abstract
Diabetic retinopathy (DR) is the most common complication of diabetes mellitus (DM), which can lead to visual impairment and even blindness in severe cases. DR is generally considered to be a microvascular disease but its pathogenesis is still unclear. A large body of evidence shows that the development of DR is not determined by a single factor but rather by multiple related mechanisms that lead to different degrees of retinal damage in DR patients. Therefore, this article briefly reviews the pathophysiological changes in DR, and discusses the occurrence and development of DR resulting from different factors such as oxidative stress, inflammation, neovascularization, neurodegeneration, the neurovascular unit, and gut microbiota, to provide a theoretical reference for the development of new DR treatment strategies.
Collapse
Affiliation(s)
- Lindan Wei
- Special Key Laboratory of Ocular Diseases of Guizhou Province, Department of Immunology, Zunyi Medical University, Zunyi, China
| | - Xin Sun
- Special Key Laboratory of Gene Detection and Therapy of Guizhou Province, School of Basic Medical Sciences, Zunyi Medical University, Zunyi, China
| | - Chenxi Fan
- Special Key Laboratory of Ocular Diseases of Guizhou Province, Department of Immunology, Zunyi Medical University, Zunyi, China
| | - Rongli Li
- Special Key Laboratory of Ocular Diseases of Guizhou Province, Department of Immunology, Zunyi Medical University, Zunyi, China
| | - Shuanglong Zhou
- Special Key Laboratory of Ocular Diseases of Guizhou Province, Department of Immunology, Zunyi Medical University, Zunyi, China
| | - Hongsong Yu
- Special Key Laboratory of Ocular Diseases of Guizhou Province, Department of Immunology, Zunyi Medical University, Zunyi, China
- *Correspondence: Hongsong Yu,
| |
Collapse
|
17
|
Veldsman T, Swanepoel M, Monyeki MA, Brits JS, Malan L. The Role of Physical Activity Status in the Relationship between Obesity and Carotid Intima-Media Thickness (CIMT) in Urban South African Teachers: The SABPA Study. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2022; 19:6348. [PMID: 35627885 PMCID: PMC9141222 DOI: 10.3390/ijerph19106348] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Revised: 05/13/2022] [Accepted: 05/18/2022] [Indexed: 02/01/2023]
Abstract
Globally, the prevalence of physical inactivity and obesity are on the rise, which may increase carotid intima-media thickness (CIMT) as a marker of subclinical atherosclerosis. This study assessed the association between physical activity (PA), obesity, and CIMT. A cross-sectional study design was used, including a sub-sample (n = 216) of teachers who participated in the Sympathetic Activity and Ambulatory Blood Pressure in Africans (SAPBA) study. Measurements included the following: physical activity status (measured with ActiHeart devices over 7 consecutive days), body mass index (BMI), waist circumference (WC), waist-to-height ratio (WtHR), CIMT (measured by SonoSite Micromax ultrasound), blood pressure (BP), fasting C-reactive protein (CRP), and cholesterol and glucose levels. Data were analysed using the Statistical Package for Social Science. One-third of the teachers were physically inactive (33%) and had low-grade inflammation CRP ≥ 3 mg/L (41%). Males were more sedentary and had higher BP and CIMT (p < 0.05). Independent of age and sex, WC or central obesity was 2.63 times more likely (p = 0.02) to contribute to atherosclerosis, especially in females (OR: 4.23, p = 0.04). PA levels were insignificantly and negatively (β −0.034; 0.888; 0.240) related to subclinical atherosclerosis. The cardiovascular disease risk profiles and limited PA status may have curbed the beneficial impact of PA on the obesity and atherosclerosis.
Collapse
Affiliation(s)
- Tamrin Veldsman
- Physical Activity, Sport and Recreation Research Focus Area (PhASRec), Faculty of Health Sciences, North-West University, Private Bag X6001, Potchefstroom 2520, South Africa; (M.S.); (M.A.M.)
| | - Mariette Swanepoel
- Physical Activity, Sport and Recreation Research Focus Area (PhASRec), Faculty of Health Sciences, North-West University, Private Bag X6001, Potchefstroom 2520, South Africa; (M.S.); (M.A.M.)
| | - Makama Andries Monyeki
- Physical Activity, Sport and Recreation Research Focus Area (PhASRec), Faculty of Health Sciences, North-West University, Private Bag X6001, Potchefstroom 2520, South Africa; (M.S.); (M.A.M.)
| | - Johanna Susanna Brits
- Hypertension in Africa Research Team (HART), North-West University, Private Bag X6001, Potchefstroom 2520, South Africa;
| | - Leoné Malan
- Technology Transfer and Innovation-Support Office, North-West University, Private Bag X1290, Potchefstroom 2520, South Africa;
| |
Collapse
|
18
|
Characterization of an Immortalized Human Microglial Cell Line as a Tool for the Study of Diabetic Retinopathy. Int J Mol Sci 2022; 23:ijms23105745. [PMID: 35628555 PMCID: PMC9145666 DOI: 10.3390/ijms23105745] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Revised: 05/12/2022] [Accepted: 05/19/2022] [Indexed: 12/04/2022] Open
Abstract
The complexity of the retinal structure reflects on the difficulty to describe its composite cell interactions. Microglia is responsible for the immune reaction to inflammatory stimuli during diabetic retinopathy (DR), but most studies still use rodent cells. We characterized a commercially available immortalized human microglial line and tested its susceptibility to inflammation, to study the interactions between the neuro-vascular retinal portions in species-specific models. After checking the expression of microglial markers, we tried lipopolysaccharide (LPS) stimulation and several pro-inflammatory cocktails to select the best combination able to induce a significant M1 (inflammatory) response. We measured M1 induction through the expression of pro- and anti-inflammatory molecules and performed morphologic and functional assays. Marker expression confirmed the human microglial derivation of these cells. Differently from rodents, LPS did not induce a M1 profile. The best pro-inflammatory stimulus was an interleukin-1β + tumor necrosis factor-α + interferon-γ cocktail, which induced morphology changes and increased proliferation, apoptosis, migration, reactive oxygen species, and the expression of inflammatory cytokines and miRNAs. In conclusion, this microglial line proved potentially useful to investigate the cascade of events leading to DR. In perspective, co-culture models involving microvascular cells will help in the understanding of multifaceted interactions of the neurovascular unit.
Collapse
|
19
|
Wareham LK, Liddelow SA, Temple S, Benowitz LI, Di Polo A, Wellington C, Goldberg JL, He Z, Duan X, Bu G, Davis AA, Shekhar K, Torre AL, Chan DC, Canto-Soler MV, Flanagan JG, Subramanian P, Rossi S, Brunner T, Bovenkamp DE, Calkins DJ. Solving neurodegeneration: common mechanisms and strategies for new treatments. Mol Neurodegener 2022; 17:23. [PMID: 35313950 PMCID: PMC8935795 DOI: 10.1186/s13024-022-00524-0] [Citation(s) in RCA: 109] [Impact Index Per Article: 54.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Accepted: 02/18/2022] [Indexed: 02/06/2023] Open
Abstract
Across neurodegenerative diseases, common mechanisms may reveal novel therapeutic targets based on neuronal protection, repair, or regeneration, independent of etiology or site of disease pathology. To address these mechanisms and discuss emerging treatments, in April, 2021, Glaucoma Research Foundation, BrightFocus Foundation, and the Melza M. and Frank Theodore Barr Foundation collaborated to bring together key opinion leaders and experts in the field of neurodegenerative disease for a virtual meeting titled "Solving Neurodegeneration". This "think-tank" style meeting focused on uncovering common mechanistic roots of neurodegenerative disease and promising targets for new treatments, catalyzed by the goal of finding new treatments for glaucoma, the world's leading cause of irreversible blindness and the common interest of the three hosting foundations. Glaucoma, which causes vision loss through degeneration of the optic nerve, likely shares early cellular and molecular events with other neurodegenerative diseases of the central nervous system. Here we discuss major areas of mechanistic overlap between neurodegenerative diseases of the central nervous system: neuroinflammation, bioenergetics and metabolism, genetic contributions, and neurovascular interactions. We summarize important discussion points with emphasis on the research areas that are most innovative and promising in the treatment of neurodegeneration yet require further development. The research that is highlighted provides unique opportunities for collaboration that will lead to efforts in preventing neurodegeneration and ultimately vision loss.
Collapse
Affiliation(s)
- Lauren K Wareham
- Department of Ophthalmology and Visual Sciences, Vanderbilt Eye Institute, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Shane A Liddelow
- Neuroscience Institute, NYU Grossman School of Medicine, New York, NY, USA
| | - Sally Temple
- Neural Stem Cell Institute, NY, 12144, Rensselaer, USA
| | - Larry I Benowitz
- Department of Neurosurgery and F.M. Kirby Neurobiology Center, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Adriana Di Polo
- Department of Neuroscience, University of Montreal, Montreal, QC, Canada
| | - Cheryl Wellington
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Jeffrey L Goldberg
- Spencer Center for Vision Research, Byers Eye Institute, Stanford University, CA, Palo Alto, USA
| | - Zhigang He
- F.M. Kirby Neurobiology Center, Boston Children's Hospital, MA, Boston, USA
| | - Xin Duan
- Department of Ophthalmology, University of California San Francisco, San Francisco, CA, USA
| | - Guojun Bu
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, USA
| | - Albert A Davis
- Department of Neurology, Washington University in St. Louis, St. Louis, Missouri, USA
| | - Karthik Shekhar
- Department of Chemical and Biomolecular Engineering and Helen Wills Neuroscience Institute, University of California Berkeley, Berkeley, CA, USA
| | - Anna La Torre
- Department of Cell Biology and Human Anatomy, University of California Davis, Davis, CA, USA
| | - David C Chan
- Division of Biology and Biological Engineering, California Institute of Technology, CA, 91125, Pasadena, USA
| | - M Valeria Canto-Soler
- CellSight Ocular Stem Cell and Regeneration Research Program, Department of Ophthalmology, Sue Anschutz-Rodgers Eye Center, University of Colorado, Aurora, CO, USA
| | - John G Flanagan
- Herbert Wertheim School of Optometry and Vision Science, University of California Berkeley, Berkeley, CA, USA
| | | | | | | | | | - David J Calkins
- Department of Ophthalmology and Visual Sciences, Vanderbilt Eye Institute, Vanderbilt University Medical Center, Nashville, TN, USA.
| |
Collapse
|
20
|
Miller LR, Tarantini S, Nyúl-Tóth Á, Johnston MP, Martin T, Bullen EC, Bickel MA, Sonntag WE, Yabluchanskiy A, Csiszar A, Ungvari ZI, Elliott MH, Conley SM. Increased Susceptibility to Cerebral Microhemorrhages Is Associated With Imaging Signs of Microvascular Degeneration in the Retina in an Insulin-Like Growth Factor 1 Deficient Mouse Model of Accelerated Aging. Front Aging Neurosci 2022; 14:788296. [PMID: 35356301 PMCID: PMC8959924 DOI: 10.3389/fnagi.2022.788296] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2021] [Accepted: 01/31/2022] [Indexed: 01/21/2023] Open
Abstract
Age-related cerebrovascular defects contribute to vascular cognitive impairment and dementia (VCID) as well as other forms of dementia. There has been great interest in developing biomarkers and other tools for studying cerebrovascular disease using more easily accessible tissues outside the brain such as the retina. Decreased circulating insulin-like growth factor 1 (IGF-1) levels in aging are thought to contribute to the development of cerebrovascular impairment, a hypothesis that has been supported by the use of IGF-1 deficient animal models. Here we evaluate vascular and other retinal phenotypes in animals with circulating IGF-1 deficiency and ask whether the retina mimics common age-related vascular changes in the brain such as the development of microhemorrhages. Using a hypertension-induced model, we confirm that IGF-1 deficient mice exhibited worsened microhemorrhages than controls. The retinas of IGF-1 deficient animals do not exhibit microhemorrhages but do exhibit signs of vascular damage and retinal stress such as patterns of vascular constriction and Müller cell activation. These signs of retinal stress are not accompanied by retinal degeneration or impaired neuronal function. These data suggest that the role of IGF-1 in the retina is complex, and while IGF-1 deficiency leads to vascular defects in both the brain and the retina, not all brain pathologies are evident in the retina.
Collapse
Affiliation(s)
- Lauren R. Miller
- Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
| | - Stefano Tarantini
- Vascular Cognitive Impairment and Neurodegeneration Program, Oklahoma Center for Geroscience and Healthy Brain Aging, Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
- Stephenson Cancer Center, University of Oklahoma, Oklahoma City, OK, United States
- International Training Program in Geroscience, Doctoral School of Basic and Translational Medicine/Department of Public Health, Semmelweis University, Budapest, Hungary
| | - Ádám Nyúl-Tóth
- Vascular Cognitive Impairment and Neurodegeneration Program, Oklahoma Center for Geroscience and Healthy Brain Aging, Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
- International Training Program in Geroscience, Doctoral School of Basic and Translational Medicine/Department of Public Health, Semmelweis University, Budapest, Hungary
- International Training Program in Geroscience, Institute of Biophysics, Biological Research Centre, Eötvös Loránd Research Network, Szeged, Hungary
| | - Morgan P. Johnston
- Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
| | - Teryn Martin
- Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
| | - Elizabeth C. Bullen
- Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
| | - Marisa A. Bickel
- Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
| | - William E. Sonntag
- Vascular Cognitive Impairment and Neurodegeneration Program, Oklahoma Center for Geroscience and Healthy Brain Aging, Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
| | - Andriy Yabluchanskiy
- Vascular Cognitive Impairment and Neurodegeneration Program, Oklahoma Center for Geroscience and Healthy Brain Aging, Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
| | - Anna Csiszar
- Vascular Cognitive Impairment and Neurodegeneration Program, Oklahoma Center for Geroscience and Healthy Brain Aging, Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
- International Training Program in Geroscience, Doctoral School of Basic and Translational Medicine/Department of Public Health, Semmelweis University, Budapest, Hungary
| | - Zoltan I. Ungvari
- Vascular Cognitive Impairment and Neurodegeneration Program, Oklahoma Center for Geroscience and Healthy Brain Aging, Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
- International Training Program in Geroscience, Doctoral School of Basic and Translational Medicine/Department of Public Health, Semmelweis University, Budapest, Hungary
| | - Michael H. Elliott
- Department of Ophthalmology, Dean McGee Eye Institute, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
- Department of Physiology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
| | - Shannon M. Conley
- Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
| |
Collapse
|
21
|
Roy A, Tolone A, Hilhorst R, Groten J, Tomar T, Paquet-Durand F. Kinase activity profiling identifies putative downstream targets of cGMP/PKG signaling in inherited retinal neurodegeneration. Cell Death Dis 2022; 8:93. [PMID: 35241647 PMCID: PMC8894370 DOI: 10.1038/s41420-022-00897-7] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Revised: 12/16/2021] [Accepted: 02/08/2022] [Indexed: 11/15/2022]
Abstract
Inherited retinal diseases (IRDs) are a group of neurodegenerative disorders that lead to photoreceptor cell death and eventually blindness. IRDs are characterised by a high genetic heterogeneity, making it imperative to design mutation-independent therapies. Mutations in a number of IRD disease genes have been associated with a rise of cyclic 3’,5’-guanosine monophosphate (cGMP) levels in photoreceptors. Accordingly, the cGMP-dependent protein kinase (PKG) has emerged as a new potential target for the mutation-independent treatment of IRDs. However, the substrates of PKG and the downstream degenerative pathways triggered by its activity have yet to be determined. Here, we performed kinome activity profiling of different murine organotypic retinal explant cultures (diseased rd1 and wild-type controls) using multiplex peptide microarrays to identify proteins whose phosphorylation was significantly altered by PKG activity. In addition, we tested the downstream effect of a known PKG inhibitor CN03 in these organotypic retina cultures. Among the PKG substrates were potassium channels belonging to the Kv1 family (KCNA3, KCNA6), cyclic AMP-responsive element-binding protein 1 (CREB1), DNA topoisomerase 2-α (TOP2A), 6-phosphofructo-2-kinase/fructose-2,6-biphosphatase 3 (F263), and the glutamate ionotropic receptor kainate 2 (GRIK2). The retinal expression of these PKG targets was further confirmed by immunofluorescence and could be assigned to various neuronal cell types, including photoreceptors, horizontal cells, and ganglion cells. Taken together, this study confirmed the key role of PKG in photoreceptor cell death and identified new downstream targets of cGMP/PKG signalling that will improve the understanding of the degenerative mechanisms underlying IRDs.
Collapse
Affiliation(s)
- Akanksha Roy
- Division of Toxicology, Wageningen University and Research, 96708 WE, Wageningen, The Netherlands.,PamGene International B.V, 5200 BJ, s-Hertogenbosch, The Netherlands
| | - Arianna Tolone
- Cell Death Mechanism Group, Institute for Ophthalmic Research, Eberhard-Karls-Universität, Tübingen, 72072, Germany
| | - Riet Hilhorst
- PamGene International B.V, 5200 BJ, s-Hertogenbosch, The Netherlands
| | - John Groten
- Division of Toxicology, Wageningen University and Research, 96708 WE, Wageningen, The Netherlands.,PamGene International B.V, 5200 BJ, s-Hertogenbosch, The Netherlands
| | - Tushar Tomar
- PamGene International B.V, 5200 BJ, s-Hertogenbosch, The Netherlands.
| | - François Paquet-Durand
- Cell Death Mechanism Group, Institute for Ophthalmic Research, Eberhard-Karls-Universität, Tübingen, 72072, Germany.
| |
Collapse
|
22
|
Kaur G, Singh NK. The Role of Inflammation in Retinal Neurodegeneration and Degenerative Diseases. Int J Mol Sci 2021; 23:ijms23010386. [PMID: 35008812 PMCID: PMC8745623 DOI: 10.3390/ijms23010386] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 12/24/2021] [Accepted: 12/28/2021] [Indexed: 12/19/2022] Open
Abstract
Retinal neurodegeneration is predominantly reported as the apoptosis or impaired function of the photoreceptors. Retinal degeneration is a major causative factor of irreversible vision loss leading to blindness. In recent years, retinal degenerative diseases have been investigated and many genes and genetic defects have been elucidated by many of the causative factors. An enormous amount of research has been performed to determine the pathogenesis of retinal degenerative conditions and to formulate the treatment modalities that are the critical requirements in this current scenario. Encouraging results have been obtained using gene therapy. We provide a narrative review of the various studies performed to date on the role of inflammation in human retinal degenerative diseases such as age-related macular degeneration, inherited retinal dystrophies, retinitis pigmentosa, Stargardt macular dystrophy, and Leber congenital amaurosis. In addition, we have highlighted the pivotal role of various inflammatory mechanisms in the progress of retinal degeneration. This review also offers an assessment of various therapeutic approaches, including gene-therapies and stem-cell-based therapies, for degenerative retinal diseases.
Collapse
Affiliation(s)
- Geetika Kaur
- Integrative Biosciences Center, Wayne State University, Detroit, MI 48202, USA;
- Department of Ophthalmology, Visual and Anatomical Sciences, Wayne State University School of Medicine, Detroit, MI 48202, USA
| | - Nikhlesh K. Singh
- Integrative Biosciences Center, Wayne State University, Detroit, MI 48202, USA;
- Department of Ophthalmology, Visual and Anatomical Sciences, Wayne State University School of Medicine, Detroit, MI 48202, USA
- Correspondence:
| |
Collapse
|
23
|
Loss of αA or αB-Crystallin Accelerates Photoreceptor Cell Death in a Mouse Model of P23H Autosomal Dominant Retinitis Pigmentosa. Int J Mol Sci 2021; 23:ijms23010070. [PMID: 35008496 PMCID: PMC8744961 DOI: 10.3390/ijms23010070] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 12/17/2021] [Accepted: 12/20/2021] [Indexed: 02/08/2023] Open
Abstract
Inherited retinal degenerations (IRD) are a leading cause of visual impairment and can result from mutations in any one of a multitude of genes. Mutations in the light-sensing protein rhodopsin (RHO) is a leading cause of IRD with the most common of those being a missense mutation that results in substitution of proline-23 with histidine. This variant, also known as P23H-RHO, results in rhodopsin misfolding, initiation of endoplasmic reticulum stress, the unfolded protein response, and activation of cell death pathways. In this study, we investigate the effect of α-crystallins on photoreceptor survival in a mouse model of IRD secondary to P23H-RHO. We find that knockout of either αA- or αB-crystallin results in increased intraretinal inflammation, activation of apoptosis and necroptosis, and photoreceptor death. Our data suggest an important role for the ⍺-crystallins in regulating photoreceptor survival in the P23H-RHO mouse model of IRD.
Collapse
|
24
|
Gong P, Zou Y, Zhang W, Tian Q, Han S, Xu Z, Chen Q, Wang X, Li M. The neuroprotective effects of Insulin-Like Growth Factor 1 via the Hippo/YAP signaling pathway are mediated by the PI3K/AKT cascade following cerebral ischemia/reperfusion injury. Brain Res Bull 2021; 177:373-387. [PMID: 34717965 DOI: 10.1016/j.brainresbull.2021.10.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2021] [Revised: 10/07/2021] [Accepted: 10/26/2021] [Indexed: 10/20/2022]
Abstract
Insulin-like growth factor 1 (IGF-1) has neuroprotective actions, including vasodilatory, anti-inflammatory, and antithrombotic effects, following ischemic stroke. However, the molecular mechanisms underlying the neuroprotective effects of IGF-1 following ischemic stroke remain unknown. Therefore, in the present study, we investigated whether IGF-1 exerted its neuroprotective effects by regulating the Hippo/YAP signaling pathway, potentially via activation of the PI3K/AKT cascade, following ischemic stroke. In the in vitro study, we exposed cultured PC12 and SH-5YSY cells, and cortical primary neurons, to oxygen-glucose deprivation. Cell viability was measured using CCK-8 assay. In the in vivo study, Sprague-Dawley rats were subjected to middle cerebral artery occlusion. Neurological function was assessed using a modified neurologic scoring system and the modified neurological severity score (mNSS) test, brain edema was detected by brain water content measurement, infarct volume was measured using triphenyltetrazolium chloride staining, and neuronal death and apoptosis were evaluated by TUNEL/NeuN double staining, HE and Nissl staining, and immunohistochemistry staining for NeuN. Finally, western blot analysis was used to measure the level of IGF-1 in vivo and levels of YAP/TAZ, PI3K and phosphorylated AKT (p-AKT) both in vitro and in vivo. IGF-1 induced activation of YAP/TAZ, which resulted in improved cell viability in vitro, and reduced neurological deficits, brain water content, neuronal death and apoptosis, and cerebral infarct volume in vivo. Notably, the neuroprotective effects of IGF-1 were blocked by an inhibitor of the PI3K/AKT cascade, LY294002. LY294002 treatment not only downregulated PI3K and p-AKT, but YAP/TAZ as well, leading to aggravation of neurological dysfunction and worsening of brain damage. Our findings indicate that the neuroprotective effects of IGF-1 are, at least in part mediated by upregulation of YAP/TAZ via activation of the PI3K/AKT cascade following cerebral ischemic stroke.
Collapse
Affiliation(s)
- Pian Gong
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, Hubei, 430060, PR China
| | - Yichun Zou
- Department of Neurosurgery, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, 430071, PR China
| | - Wei Zhang
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, Hubei, 430060, PR China
| | - Qi Tian
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, Hubei, 430060, PR China
| | - Shoumeng Han
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, Hubei, 430060, PR China
| | - Zhou Xu
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, Hubei, 430060, PR China
| | - Qianxue Chen
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, Hubei, 430060, PR China
| | - Xin Wang
- Department of Neurosurgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Mingchang Li
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, Hubei, 430060, PR China.
| |
Collapse
|
25
|
Meng C, Gu C, He S, Su T, Lhamo T, Draga D, Qiu Q. Pyroptosis in the Retinal Neurovascular Unit: New Insights Into Diabetic Retinopathy. Front Immunol 2021; 12:763092. [PMID: 34737754 PMCID: PMC8560732 DOI: 10.3389/fimmu.2021.763092] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Accepted: 09/28/2021] [Indexed: 12/15/2022] Open
Abstract
Diabetic retinopathy (DR) is prevalent among people with long-term diabetes mellitus (DM) and remains the leading cause of visual impairment in working-aged people. DR is related to chronic low-level inflammatory reactions. Pyroptosis is an emerging type of inflammatory cell death mediated by gasdermin D (GSDMD), NOD-like receptors and inflammatory caspases that promote interleukin-1β (IL-1β) and IL-18 release. In addition, the retinal neurovascular unit (NVU) is the functional basis of the retina. Recent studies have shown that pyroptosis may participate in the destruction of retinal NVU cells in simulated hyperglycemic DR environments. In this review, we will clarify the importance of pyroptosis in the retinal NVU during the development of DR.
Collapse
Affiliation(s)
- Chunren Meng
- Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- National Clinical Research Center for Eye Diseases; Shanghai Key Laboratory of Ocular Fundus Diseases, Shanghai Engineering Center for Visual Science and Photomedicine, Shanghai Engineering Center for Precise Diagnosis and Treatment of Eye Diseases, Shanghai, China
| | - Chufeng Gu
- Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- National Clinical Research Center for Eye Diseases; Shanghai Key Laboratory of Ocular Fundus Diseases, Shanghai Engineering Center for Visual Science and Photomedicine, Shanghai Engineering Center for Precise Diagnosis and Treatment of Eye Diseases, Shanghai, China
| | - Shuai He
- Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- National Clinical Research Center for Eye Diseases; Shanghai Key Laboratory of Ocular Fundus Diseases, Shanghai Engineering Center for Visual Science and Photomedicine, Shanghai Engineering Center for Precise Diagnosis and Treatment of Eye Diseases, Shanghai, China
| | - Tong Su
- Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- National Clinical Research Center for Eye Diseases; Shanghai Key Laboratory of Ocular Fundus Diseases, Shanghai Engineering Center for Visual Science and Photomedicine, Shanghai Engineering Center for Precise Diagnosis and Treatment of Eye Diseases, Shanghai, China
| | - Thashi Lhamo
- Department of Ophthalmology, Shigatse People’s Hospital, Shigatse, China
| | - Deji Draga
- Department of Ophthalmology, Shigatse People’s Hospital, Shigatse, China
| | - Qinghua Qiu
- Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- National Clinical Research Center for Eye Diseases; Shanghai Key Laboratory of Ocular Fundus Diseases, Shanghai Engineering Center for Visual Science and Photomedicine, Shanghai Engineering Center for Precise Diagnosis and Treatment of Eye Diseases, Shanghai, China
- Department of Ophthalmology, Shigatse People’s Hospital, Shigatse, China
| |
Collapse
|
26
|
Li X, Cai S, He Z, Reilly J, Zeng Z, Strang N, Shu X. Metabolomics in Retinal Diseases: An Update. BIOLOGY 2021; 10:944. [PMID: 34681043 PMCID: PMC8533136 DOI: 10.3390/biology10100944] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Revised: 09/16/2021] [Accepted: 09/18/2021] [Indexed: 12/17/2022]
Abstract
Retinal diseases are a leading cause of visual loss and blindness, affecting a significant proportion of the population worldwide and having a detrimental impact on quality of life, with consequent economic burden. The retina is highly metabolically active, and a number of retinal diseases are associated with metabolic dysfunction. To better understand the pathogenesis underlying such retinopathies, new technology has been developed to elucidate the mechanism behind retinal diseases. Metabolomics is a relatively new "omics" technology, which has developed subsequent to genomics, transcriptomics, and proteomics. This new technology can provide qualitative and quantitative information about low-molecular-weight metabolites (M.W. < 1500 Da) in a given biological system, which shed light on the physiological or pathological state of a cell or tissue sample at a particular time point. In this article we provide an extensive review of the application of metabolomics to retinal diseases, with focus on age-related macular degeneration (AMD), diabetic retinopathy (DR), retinopathy of prematurity (ROP), glaucoma, and retinitis pigmentosa (RP).
Collapse
Affiliation(s)
- Xing Li
- School of Basic Medical Sciences, Shaoyang University, Shaoyang 422000, China; (X.L.); (Z.H.)
| | - Shichang Cai
- Department of Human Anatomy, School of Medicine, Hunan University of Medicine, Huaihua 418000, China;
| | - Zhiming He
- School of Basic Medical Sciences, Shaoyang University, Shaoyang 422000, China; (X.L.); (Z.H.)
| | - James Reilly
- Department of Biological and Biomedical Sciences, Glasgow Caledonian University, Glasgow G4 0BA, UK;
| | - Zhihong Zeng
- College of Biological and Environmental Engineering, Changsha University, Changsha 410022, China;
| | - Niall Strang
- Department of Vision Science, Glasgow Caledonian University, Glasgow G4 0BA, UK;
| | - Xinhua Shu
- School of Basic Medical Sciences, Shaoyang University, Shaoyang 422000, China; (X.L.); (Z.H.)
- Department of Biological and Biomedical Sciences, Glasgow Caledonian University, Glasgow G4 0BA, UK;
- Department of Vision Science, Glasgow Caledonian University, Glasgow G4 0BA, UK;
| |
Collapse
|
27
|
Campello L, Singh N, Advani J, Mondal AK, Corso-Díaz X, Swaroop A. Aging of the Retina: Molecular and Metabolic Turbulences and Potential Interventions. Annu Rev Vis Sci 2021; 7:633-664. [PMID: 34061570 PMCID: PMC11375453 DOI: 10.1146/annurev-vision-100419-114940] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Multifaceted and divergent manifestations across tissues and cell types have curtailed advances in deciphering the cellular events that accompany advanced age and contribute to morbidities and mortalities. Increase in human lifespan during the past century has heightened awareness of the need to prevent age-associated frailty of neuronal and sensory systems to allow a healthy and productive life. In this review, we discuss molecular and physiological attributes of aging of the retina, with a goal of understanding age-related impairment of visual function. We highlight the epigenome-metabolism nexus and proteostasis as key contributors to retinal aging and discuss lifestyle changes as potential modulators of retinal function. Finally, we deliberate promising intervention strategies for promoting healthy aging of the retina for improved vision.
Collapse
Affiliation(s)
- Laura Campello
- Neurobiology, Neurodegeneration and Repair Laboratory, National Eye Institute, National Institutes of Health, Bethesda, Maryland 20892, USA;
| | - Nivedita Singh
- Neurobiology, Neurodegeneration and Repair Laboratory, National Eye Institute, National Institutes of Health, Bethesda, Maryland 20892, USA;
| | - Jayshree Advani
- Neurobiology, Neurodegeneration and Repair Laboratory, National Eye Institute, National Institutes of Health, Bethesda, Maryland 20892, USA;
| | - Anupam K Mondal
- Neurobiology, Neurodegeneration and Repair Laboratory, National Eye Institute, National Institutes of Health, Bethesda, Maryland 20892, USA;
| | - Ximena Corso-Díaz
- Neurobiology, Neurodegeneration and Repair Laboratory, National Eye Institute, National Institutes of Health, Bethesda, Maryland 20892, USA;
| | - Anand Swaroop
- Neurobiology, Neurodegeneration and Repair Laboratory, National Eye Institute, National Institutes of Health, Bethesda, Maryland 20892, USA;
| |
Collapse
|
28
|
Kovács-Valasek A, Pöstyéni E, Dénes V, Mester A, Sétáló G, Gábriel R. Age-Related Alterations of Proteins in Albino Wistar Rat Retina. Cells Tissues Organs 2021; 210:135-150. [PMID: 34218223 DOI: 10.1159/000515447] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Accepted: 02/04/2021] [Indexed: 01/05/2023] Open
Abstract
Imbalance of homeostasis causes permanent changes in the body with time. The central nervous system is especially prone to these changes since it possesses limited regenerative capacity. In the retina, neurons are damaged during the aging process, and this eventually leads to deterioration of vision. In our 2-year-long study, we examined genetically closely related rat individuals to disclose the hidden retinal causes of age-associated visual dysfunction. Morphometric analysis showed significant reduction of the retina thickness with aging, particularly that of the inner plexiform layer. To reveal changes between the age groups, we used immunohistochemistry against vesicular glutamate transporter 1 protein for photoreceptor and bipolar cell terminals, Brn3a for ganglion cells, calbindin 28 kDa for horizontal cells, parvalbumin for AII amacrines, protein kinase Cα for rod bipolar cells, tyrosine hydroxylase for dopaminergic cells, glial fibrillary acidic protein for glial cells, and peanut-agglutinin labeling for cones. The most significant decrease was observed in the density of photoreceptor and the ganglion cells in the aging process. By using immunocytochemistry and western blot technique, we observed that calbindin and vesicular glutamate transporter 1 protein staining do not change much with aging; tyrosine hydroxylase, parvalbumin and calretinin showed the highest immunoreactivity during the midlife period. Most interestingly, the level of glial fibrillary acidic protein also changes similarly to the previously named markers. Our results provide further evidence that protein content is modified at least in some cell populations of the rat retina, and the number of retinal cells declined with aging. We conclude that senescence alone may cause structural and functional damage in the retinal tissue.
Collapse
Affiliation(s)
- Andrea Kovács-Valasek
- Department of Experimental Zoology and Neurobiology, Institute of Biology, Faculty of Sciences, University of Pécs, Pécs, Hungary.,János Szentágothai Research Centre, University of Pécs, Pécs, Hungary
| | - Etelka Pöstyéni
- Department of Experimental Zoology and Neurobiology, Institute of Biology, Faculty of Sciences, University of Pécs, Pécs, Hungary
| | - Viktória Dénes
- Department of Experimental Zoology and Neurobiology, Institute of Biology, Faculty of Sciences, University of Pécs, Pécs, Hungary
| | - Adrienn Mester
- Department of Experimental Zoology and Neurobiology, Institute of Biology, Faculty of Sciences, University of Pécs, Pécs, Hungary
| | - György Sétáló
- Department of Medical Biology, Medical School, University of Pécs, Pécs, Hungary.,János Szentágothai Research Centre, University of Pécs, Pécs, Hungary
| | - Róbert Gábriel
- Department of Experimental Zoology and Neurobiology, Institute of Biology, Faculty of Sciences, University of Pécs, Pécs, Hungary.,János Szentágothai Research Centre, University of Pécs, Pécs, Hungary
| |
Collapse
|
29
|
Delivery Systems of Retinoprotective Proteins in the Retina. Int J Mol Sci 2021; 22:ijms22105344. [PMID: 34069505 PMCID: PMC8160820 DOI: 10.3390/ijms22105344] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Revised: 05/12/2021] [Accepted: 05/17/2021] [Indexed: 12/26/2022] Open
Abstract
Retinoprotective proteins play important roles for retinal tissue integrity. They can directly affect the function and the survival of photoreceptors, and/or indirectly target the retinal pigment epithelium (RPE) and endothelial cells that support these tissues. Retinoprotective proteins are used in basic, translational and in clinical studies to prevent and treat human retinal degenerative disorders. In this review, we provide an overview of proteins that protect the retina and focus on pigment epithelium-derived factor (PEDF), and its effects on photoreceptors, RPE cells, and endothelial cells. We also discuss delivery systems such as pharmacologic and genetic administration of proteins to achieve photoreceptor survival and retinal tissue integrity.
Collapse
|
30
|
Rettinger CL, Kaini RR, Burke TA, Wang HC. Neurotrophic Factors Secreted by Induced Pluripotent Stem Cell-Derived Retinal Progenitors Promote Retinal Survival and Preservation in an Adult Porcine Neuroretina Model. J Ocul Pharmacol Ther 2021; 37:301-312. [PMID: 33661042 DOI: 10.1089/jop.2020.0088] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Purpose: Paracrine factors released by pluripotent stem cells have shown great potential as therapeutic agents in regenerative medicine. The purpose of this study was to characterize trophic factor secretion of retinal progenitor cells (RPCs) derived from human induced pluripotent stem cells (iPSCs) and to assess its impact on retinal survival ex vivo. Methods: RPCs were generated from human 3D1 iPSCs following previously established protocols with modifications. Conditioned medium (CM) was harvested from iPSC-derived retinal progenitors and analyzed for trophic factor composition through multiplex enzyme-linked immunosorbent assay. Retina-preserving capability of the collected CM was examined using a degenerative porcine neuroretina model. Viability of the CM-treated retina explants was evaluated using the resazurin-based PrestoBlue reagent, whereas the lactate dehydrogenase (LDH) assay was used to assess retinal cytotoxicity. Retina explants were also analyzed morphologically through immunohistochemistry for glial cell activation and apoptosis. Results: We have successfully generated and characterized iPSC-derived RPCs that secreted an array of neuroprotective factors, including osteopontin, hepatocyte growth factor, stromal cell-derived factor 1, and insulin-like growth factor-1. Retina explants cultured in CM derived from iPSC-RPCs (iPSC-RPC-CM) showed better preservation of the retinal microarchitecture and fewer terminal deoxynucleotidyl transferase dUTP nick-end labeling (TUNEL)+ nuclei, and reduced reactive gliosis. Furthermore, we saw a reduction in extracellular LDH levels in CM-treated retina explants, which also exhibited higher metabolic activity than the untreated controls. Conclusions: iPSC-derived RPCs secrete many trophic factors that have been shown to promote neuroprotection, tissue repair, and regeneration in the retina. Overall, we have demonstrated the neuroprotective effects of iPSC-RPC-CM through a degenerative neuroretina model ex vivo.
Collapse
Affiliation(s)
- Christina L Rettinger
- Ocular and Sensory Trauma Task Area, United States Army Institute of Surgical Research, Fort Sam Houston, Texas, USA
| | - Ramesh R Kaini
- Ocular and Sensory Trauma Task Area, United States Army Institute of Surgical Research, Fort Sam Houston, Texas, USA
| | - Teresa A Burke
- Ocular and Sensory Trauma Task Area, United States Army Institute of Surgical Research, Fort Sam Houston, Texas, USA
| | - Heuy-Ching Wang
- Ocular and Sensory Trauma Task Area, United States Army Institute of Surgical Research, Fort Sam Houston, Texas, USA
| |
Collapse
|
31
|
Potilinski MC, Tate PS, Lorenc VE, Gallo JE. New insights into oxidative stress and immune mechanisms involved in age-related macular degeneration tackled by novel therapies. Neuropharmacology 2021; 188:108513. [PMID: 33662390 DOI: 10.1016/j.neuropharm.2021.108513] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Revised: 02/14/2021] [Accepted: 02/22/2021] [Indexed: 12/20/2022]
Abstract
The prevalence of age-related macular degeneration (AMD) has increased in the last years. Although anti-VEGF agents have improved the prognosis of exudative AMD, dry AMD has still devastating effects on elderly people vision. Oxidative stress and inflammation are mechanisms involved in AMD pathogenesis and its progression. Molecular pathways involving epidermal growth factor receptor (EGFR), bone morphogenetic protein (BMP4) and the nuclear erythroid related factor 2 (Nrf2) are behind oxidative stress in AMD due to their participation in antioxidant cellular pathways. As a consequence of the disbalance produced in the antioxidant mechanisms, there is an activation of innate and adaptative immune response with cell recruitment, changes in complement factors expression, and modification of cellular milieu. Different therapies are being studied to treat dry AMD based on the possible effects on antioxidant molecular pathways or their action on the immune response. There is a wide range of treatments presented in this review, from natural antioxidant compounds to cell and gene therapy, based on their mechanisms. Finally, we hypothesize that alpha-1-antitrypsin (AAT), an anti-inflammatory and immunomodulatory molecule that can also modulate antioxidant cellular defenses, could be a good candidate for testing in AMD. This article is part of the special ssue on 'The Quest for Disease-Modifying Therapies for Neurodegenerative Disorders'.
Collapse
Affiliation(s)
- María Constanza Potilinski
- Nanomedicine & Vision Lab, Instituto de Investigaciones en Medicina Translacional, Universidad Austral, CONICET, Pilar, Buenos Aires, Argentina
| | - Pablo S Tate
- Laboratorio de Enfermedades Neurodegenerativas, Instituto de Investigaciones en Medicina Translacional, Universidad Austral, CONICET, Pilar, Buenos Aires, Argentina
| | - Valeria E Lorenc
- Nanomedicine & Vision Lab, Instituto de Investigaciones en Medicina Translacional, Universidad Austral, CONICET, Pilar, Buenos Aires, Argentina
| | - Juan E Gallo
- Nanomedicine & Vision Lab, Instituto de Investigaciones en Medicina Translacional, Universidad Austral, CONICET, Pilar, Buenos Aires, Argentina; Departamento de Oftalmología, Hospital Universitario Austral, Pilar, Buenos Aires, Argentina.
| |
Collapse
|
32
|
Retinal Inflammation, Cell Death and Inherited Retinal Dystrophies. Int J Mol Sci 2021; 22:ijms22042096. [PMID: 33672611 PMCID: PMC7924201 DOI: 10.3390/ijms22042096] [Citation(s) in RCA: 56] [Impact Index Per Article: 18.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Revised: 02/12/2021] [Accepted: 02/18/2021] [Indexed: 12/15/2022] Open
Abstract
Inherited retinal dystrophies (IRDs) are a group of retinal disorders that cause progressive and severe loss of vision because of retinal cell death, mainly photoreceptor cells. IRDs include retinitis pigmentosa (RP), the most common IRD. IRDs present a genetic and clinical heterogeneity that makes it difficult to achieve proper treatment. The progression of IRDs is influenced, among other factors, by the activation of the immune cells (microglia, macrophages, etc.) and the release of inflammatory molecules such as chemokines and cytokines. Upregulation of tumor necrosis factor alpha (TNFα), a pro-inflammatory cytokine, is found in IRDs. This cytokine may influence photoreceptor cell death. Different cell death mechanisms are proposed, including apoptosis, necroptosis, pyroptosis, autophagy, excessive activation of calpains, or parthanatos for photoreceptor cell death. Some of these cell death mechanisms are linked to TNFα upregulation and inflammation. Therapeutic approaches that reduce retinal inflammation have emerged as useful therapies for slowing down the progression of IRDs. We focused this review on the relationship between retinal inflammation and the different cell death mechanisms involved in RP. We also reviewed the main anti-inflammatory therapies for the treatment of IRDs.
Collapse
|
33
|
Malan L, Hamer M, von Känel R, van Wyk RD, Sumner AE, Nilsson PM, Lambert GW, Steyn HS, Badenhorst CJ, Malan NT. A Stress Syndrome Prototype Reflects Type 3 Diabetes and Ischemic Stroke Risk: The SABPA Study. BIOLOGY 2021; 10:162. [PMID: 33670473 PMCID: PMC7922484 DOI: 10.3390/biology10020162] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Revised: 01/28/2021] [Accepted: 02/09/2021] [Indexed: 12/19/2022]
Abstract
Type 3 diabetes (T3D) accurately reflects that dementia, e.g., Alzheimer's disease, represents insulin resistance and neurodegeneration in the brain. Similar retinal microvascular changes were observed in Alzheimer's and chronic stressed individuals. Hence, we aimed to show that chronic stress relates to T3D dementia signs and retinopathy, ultimately comprising a Stress syndrome prototype reflecting risk for T3D and stroke. A chronic stress and stroke risk phenotype (Stressed) score, independent of age, race or gender, was applied to stratify participants (N = 264; aged 44 ± 9 years) into high stress risk (Stressed, N = 159) and low stress risk (non-Stressed, N = 105) groups. We determined insulin resistance using the homeostatic model assessment (HOMA-IR), which is interchangeable with T3D, and dementia risk markers (cognitive executive functioning (cognitiveexe-func); telomere length; waist circumference (WC), neuronal glia injury; neuron-specific enolase/NSE, S100B). Retinopathy was determined in the mydriatic eye. The Stressed group had greater incidence of HOMA-IR in the upper quartile (≥5), larger WC, poorer cognitiveexe-func control, shorter telomeres, consistently raised neuronal glia injury, fewer retinal arteries, narrower arteries, wider veins and a larger optic cup/disc ratio (C/D) compared to the non-Stressed group. Furthermore, of the stroke risk markers, arterial narrowing was related to glaucoma risk with a greater C/D, whilst retinal vein widening was related to HOMA-IR, poor cognitiveexe-func control and neuronal glia injury (Adjusted R2 0.30; p ≤ 0.05). These associations were not evident in the non-Stressed group. Logistic regression associations between the Stressed phenotype and four dementia risk markers (cognitiveexe-func, telomere length, NSE and WC) comprised a Stress syndrome prototype (area under the curve 0.80; sensitivity/specificity 85%/58%; p ≤ 0.001). The Stress syndrome prototype reflected risk for HOMA-IR (odds ratio (OR) 7.72) and retinal glia ischemia (OR 1.27) and vein widening (OR 1.03). The Stressed phenotype was associated with neuronal glia injury and retinal ischemia, potentiating glaucoma risk. The detrimental effect of chronic stress exemplified a Stress syndrome prototype reflecting risk for type 3 diabetes, neurodegeneration and ischemic stroke.
Collapse
Affiliation(s)
- Leoné Malan
- Hypertension in Africa Research Team (HART), North-West University, Potchefstroom 2520, South Africa; (R.v.K.); (N.T.M.)
| | - Mark Hamer
- Division of Surgery & Interventional Science, Faculty of Medical Sciences, University College London, London WC1E 6BT, UK;
| | - Roland von Känel
- Hypertension in Africa Research Team (HART), North-West University, Potchefstroom 2520, South Africa; (R.v.K.); (N.T.M.)
- Department of Consultation-Liaison Psychiatry and Psychosomatic Medicine, University Hospital Zurich, 8091 Zurich, Switzerland
| | - Roelof D. van Wyk
- Surgical Ophthalmologist, 85 Peter Mokaba Street, Potchefstroom 2531, South Africa;
| | - Anne E. Sumner
- Section on Ethnicity and Health, Diabetes, Endocrinology and Obesity Branch, National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, MD 20892, USA;
- National Institute of Minority Health and Health Disparities, National Institutes of Health, Bethesda, MD 20892, USA
| | - Peter M. Nilsson
- Department of Clinical Sciences, Lund University, SE-205 02 Malmö, Sweden;
| | - Gavin W. Lambert
- Iverson Health Innovation Research Institute, Swinburne University of Technology, Hawthorn, VIC 3122, Australia;
- Baker Heart & Diabetes Institute, Melbourne, VIC 3004, Australia
| | - Hendrik S. Steyn
- Statistical Consultation Services, North-West University, Potchefstroom 2520, South Africa;
| | - Casper J. Badenhorst
- Anglo American Corporate Services, Sustainable Development Department, Johannesburg 2017, South Africa;
| | - Nico T. Malan
- Hypertension in Africa Research Team (HART), North-West University, Potchefstroom 2520, South Africa; (R.v.K.); (N.T.M.)
| |
Collapse
|
34
|
Wu J, Gao G, Shi F, Xie H, Yang Q, Liu D, Qu S, Qin H, Zhang C, Xu GT, Liu F, Zhang J. Activated microglia-induced neuroinflammatory cytokines lead to photoreceptor apoptosis in Aβ-injected mice. J Mol Med (Berl) 2021; 99:713-728. [PMID: 33575853 DOI: 10.1007/s00109-021-02046-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Revised: 01/24/2021] [Accepted: 01/26/2021] [Indexed: 12/16/2022]
Abstract
Age-related macular degeneration (AMD) is mainly characterized by the progressive accumulation of drusen deposits and loss of photoreceptors and retinal pigment epithelial (RPE) cells. Because amyloid β (Aβ) is the main component of drusen, Aβ-induced activated microglia most likely lead to neuroinflammation and play a critical role in the pathogenesis of AMD. However, the relationship between activated microglia-mediated neuroinflammatory cytokines and photoreceptor death has not been clarified. By subretinal injection of Aβ42 in mice, we mimicked an inflammatory milieu of AMD to better understand how activated microglia-induced neuroinflammatory cytokines lead to photoreceptor apoptosis in the AMD progression. We demonstrated that subretinal injection of Aβ42 induces microglial activation and increases inflammatory cytokine release, which gives rise to photoreceptor apoptosis in mice. Our results were verified in vitro by co-culture of Aβ42 activated primary microglia and the photoreceptor cell line 661W. We also demonstrated that the p38 mitogen-activated protein kinase (MAPK) signaling pathway was involved in Aβ42-induced microglial activation and inflammatory cytokine release. Overall, our findings indicate that activated microglia-derived neuroinflammatory cytokines could contribute to photoreceptor apoptosis under the stimulation of Aβ42. Moreover, this study may provide a potential therapeutic approach for AMD. KEY MESSAGES: Further explore the association between activated microglia-derived neuroinflammatory cytokine secretion and photoreceptor apoptosis under the stimulation of Aβ42. Subretinal injection of Aβ42 induces the activation of microglia and increases proinflammatory cytokines IL-1β and COX-2 expression in the retina, which could give rise to the deterioration of visual function and aggravate photoreceptor apoptosis in mice. Primary microglial are activated and the levels of proinflammatory cytokines are increased by Aβ42 stimulation, which could increase the apoptosis of photoreceptor cell line 661W in vitro. The p38 MAPK signaling pathway is involved in microglial activation and photoreceptor apoptosis under Aβ42 treatment.
Collapse
Affiliation(s)
- Jing Wu
- Department of Ophthalmology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Ge Gao
- Center for Translational Neurodegeneration and Regenerative Therapy, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Fanjun Shi
- Department of Ophthalmology, the Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Hai Xie
- Department of Regenerative Medicine and Department of Pharmacology, Tongji University School of Medicine, Shanghai, China
| | - Qian Yang
- Department of Regenerative Medicine and Department of Pharmacology, Tongji University School of Medicine, Shanghai, China
| | - Dandan Liu
- Department of Regenerative Medicine and Department of Pharmacology, Tongji University School of Medicine, Shanghai, China
| | - Sichang Qu
- Department of Ophthalmology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Haifeng Qin
- Department of Ophthalmology, Shanghai Changhai Hospital, Shanghai, China
| | - Chaoyang Zhang
- Department of Ophthalmology, Shanghai General Hospital (Shanghai First People's Hospital), Shanghai Jiao Tong University, Shanghai, China.,National Clinical Research Center for Eye Diseases; Shanghai Key Laboratory of Ocular Fundus Diseases; Shanghai Engineering Center for Visual Science and Photomedicine, Shanghai Engineering Center for Precise Diagnosis and Treatment of Eye Diseases, Shanghai, China
| | - Guo-Tong Xu
- Department of Regenerative Medicine and Department of Pharmacology, Tongji University School of Medicine, Shanghai, China
| | - Fang Liu
- Department of Ophthalmology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China.
| | - Jingfa Zhang
- Department of Ophthalmology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China. .,Department of Regenerative Medicine and Department of Pharmacology, Tongji University School of Medicine, Shanghai, China. .,Department of Ophthalmology, Shanghai General Hospital (Shanghai First People's Hospital), Shanghai Jiao Tong University, Shanghai, China. .,National Clinical Research Center for Eye Diseases; Shanghai Key Laboratory of Ocular Fundus Diseases; Shanghai Engineering Center for Visual Science and Photomedicine, Shanghai Engineering Center for Precise Diagnosis and Treatment of Eye Diseases, Shanghai, China.
| |
Collapse
|
35
|
Lin Y, Ren X, Chen Y, Chen D. Interaction Between Mesenchymal Stem Cells and Retinal Degenerative Microenvironment. Front Neurosci 2021; 14:617377. [PMID: 33551729 PMCID: PMC7859517 DOI: 10.3389/fnins.2020.617377] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2020] [Accepted: 12/17/2020] [Indexed: 02/06/2023] Open
Abstract
Retinal degenerative diseases (RDDs) are a group of diseases contributing to irreversible vision loss with yet limited therapies. Stem cell-based therapy is a promising novel therapeutic approach in RDD treatment. Mesenchymal stromal/stem cells (MSCs) have emerged as a leading cell source due to their neurotrophic and immunomodulatory capabilities, limited ethical concerns, and low risk of tumor formation. Several pre-clinical studies have shown that MSCs have the potential to delay retinal degeneration, and recent clinical trials have demonstrated promising safety profiles for the application of MSCs in retinal disease. However, some of the clinical-stage MSC therapies have been unable to meet primary efficacy end points, and severe side effects were reported in some retinal “stem cell” clinics. In this review, we provide an update of the interaction between MSCs and the RDD microenvironment and discuss how to balance the therapeutic potential and safety concerns of MSCs' ocular application.
Collapse
Affiliation(s)
- Yu Lin
- The Research Laboratory of Ophthalmology and Vision Sciences, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China.,The Department of Ophthalmology, West China Hospital, Sichuan University, Chengdu, China
| | - Xiang Ren
- The Research Laboratory of Ophthalmology and Vision Sciences, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China.,The Department of Ophthalmology, West China Hospital, Sichuan University, Chengdu, China
| | - Yongjiang Chen
- The School of Optometry and Vision Science, University of Waterloo, Waterloo, ON, Canada
| | - Danian Chen
- The Research Laboratory of Ophthalmology and Vision Sciences, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China.,The Department of Ophthalmology, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
36
|
Blasiak J, Pawlowska E, Sobczuk A, Szczepanska J, Kaarniranta K. The Aging Stress Response and Its Implication for AMD Pathogenesis. Int J Mol Sci 2020; 21:ijms21228840. [PMID: 33266495 PMCID: PMC7700335 DOI: 10.3390/ijms21228840] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Revised: 11/12/2020] [Accepted: 11/20/2020] [Indexed: 02/07/2023] Open
Abstract
Aging induces several stress response pathways to counterbalance detrimental changes associated with this process. These pathways include nutrient signaling, proteostasis, mitochondrial quality control and DNA damage response. At the cellular level, these pathways are controlled by evolutionarily conserved signaling molecules, such as 5’AMP-activated protein kinase (AMPK), mechanistic target of rapamycin (mTOR), insulin/insulin-like growth factor 1 (IGF-1) and sirtuins, including SIRT1. Peroxisome proliferation-activated receptor coactivator 1 alpha (PGC-1α), encoded by the PPARGC1A gene, playing an important role in antioxidant defense and mitochondrial biogenesis, may interact with these molecules influencing lifespan and general fitness. Perturbation in the aging stress response may lead to aging-related disorders, including age-related macular degeneration (AMD), the main reason for vision loss in the elderly. This is supported by studies showing an important role of disturbances in mitochondrial metabolism, DDR and autophagy in AMD pathogenesis. In addition, disturbed expression of PGC-1α was shown to associate with AMD. Therefore, the aging stress response may be critical for AMD pathogenesis, and further studies are needed to precisely determine mechanisms underlying its role in AMD. These studies can include research on retinal cells produced from pluripotent stem cells obtained from AMD donors with the mutations, either native or engineered, in the critical genes for the aging stress response, including AMPK, IGF1, MTOR, SIRT1 and PPARGC1A.
Collapse
Affiliation(s)
- Janusz Blasiak
- Department of Molecular Genetics, Faculty of Biology and Environmental Protection, University of Lodz, 90-236 Lodz, Poland
- Correspondence: ; Tel.: +48-426354334
| | - Elzbieta Pawlowska
- Department of Orthodontics, Medical University of Lodz, 92-216 Lodz, Poland;
| | - Anna Sobczuk
- Department of Gynaecology and Obstetrics, Medical University of Lodz, 93-338 Lodz, Poland;
| | - Joanna Szczepanska
- Department of Pediatric Dentistry, Medical University of Lodz, 92-216 Lodz, Poland;
| | - Kai Kaarniranta
- Department of Ophthalmology, University of Eastern Finland, 70211 Kuopio, Finland;
- Department of Ophthalmology, Kuopio University Hospital, 70211 Kuopio, Finland
| |
Collapse
|
37
|
Vernazza S, Tirendi S, Bassi AM, Traverso CE, Saccà SC. Neuroinflammation in Primary Open-Angle Glaucoma. J Clin Med 2020; 9:E3172. [PMID: 33007927 PMCID: PMC7601106 DOI: 10.3390/jcm9103172] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Revised: 09/24/2020] [Accepted: 09/28/2020] [Indexed: 12/18/2022] Open
Abstract
Primary open-angle glaucoma (POAG) is the second leading cause of irreversible blindness worldwide. Increasing evidence suggests oxidative damage and immune response defects are key factors contributing to glaucoma onset. Indeed, both the failure of the trabecular meshwork tissue in the conventional outflow pathway and the neuroinflammation process, which drives the neurodegeneration, seem to be linked to the age-related over-production of free radicals (i.e., mitochondrial dysfunction) and to oxidative stress-linked immunostimulatory signaling. Several previous studies have described a wide range of oxidative stress-related makers which are found in glaucomatous patients, including low levels of antioxidant defences, dysfunction/activation of glial cells, the activation of the NF-κB pathway and the up-regulation of pro-inflammatory cytokines, and so on. However, the intraocular pressure is still currently the only risk factor modifiable by medication or glaucoma surgery. This present review aims to summarize the multiple cellular processes, which promote different risk factors in glaucoma including aging, oxidative stress, trabecular meshwork defects, glial activation response, neurodegenerative insults, and the altered regulation of immune response.
Collapse
Affiliation(s)
| | - Sara Tirendi
- Department of Experimental Medicine (DIMES), University of Genoa, 16132 Genoa, Italy; (S.T.); (A.M.B.)
- Inter-University Center for the Promotion of the 3Rs Principles in Teaching & Research (Centro 3R), Italy
| | - Anna Maria Bassi
- Department of Experimental Medicine (DIMES), University of Genoa, 16132 Genoa, Italy; (S.T.); (A.M.B.)
- Inter-University Center for the Promotion of the 3Rs Principles in Teaching & Research (Centro 3R), Italy
| | - Carlo Enrico Traverso
- Clinica Oculistica, DiNOGMI, University of Genoa, 16132 Genoa, Italy;
- Ophthalmology Unit, IRCCS-Polyclinic San Martino Hospital, 16132 Genoa, Italy;
| | | |
Collapse
|
38
|
Nahavandipour A, Krogh Nielsen M, Sørensen TL, Subhi Y. Systemic levels of interleukin-6 in patients with age-related macular degeneration: a systematic review and meta-analysis. Acta Ophthalmol 2020; 98:434-444. [PMID: 32180348 DOI: 10.1111/aos.14402] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2019] [Accepted: 02/21/2020] [Indexed: 01/14/2023]
Abstract
Age-related macular degeneration (AMD) is the most prevalent cause of irreversible vision loss in industrialized countries. Several studies have investigated systemic interleukin-6 (IL-6) levels of patients with AMD. In this study, we systemically reviewed the literature to provide an overview of the field and used meta-analyses to provide a summary estimate of the standardized mean difference (SMD) of systemic IL-6 between patients with AMD and control individuals. We searched the literature databases PubMed/MEDLINE, Embase, Web of Science and the Cochrane Central on 1 June 2019 for relevant studies on humans. Two authors independently extracted data and evaluated risk of bias. We identified 19 studies for the qualitative review with a total of more than 3586 individuals (1865 controls and 1721 with AMD). We found an overall random-effects SMD in systemic IL-6 levels 0.63 (95% CI: 0.28 to 0.99, p = 0.0005) corresponding to a medium effect size. In a subgroup analysis, we found that early AMD was not strongly associated with elevated IL-6 levels (0.12, 95% CI: -0.01 to 0.24, p = 0.06), which was in contrast to the significantly elevated IL-6 levels in patients with geographic atrophy (1.21, 95% CI: 0.41 to 2.01, p = 0.003) and patients with neovascular AMD (0.99, 95% CI: 0.34 to 1.63, p = 0.003). Our results show that the evidence today suggests an increased systemic IL-6 in patients with AMD, but that this may be a phenomenon more closely related to the late subtypes of AMD.
Collapse
Affiliation(s)
| | | | - Torben L Sørensen
- Department of Ophthalmology, Zealand University Hospital, Roskilde, Denmark.,Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Yousif Subhi
- Department of Ophthalmology, Zealand University Hospital, Roskilde, Denmark.,Department of Ophthalmology, Rigshospitalet-Glostrup, Glostrup, Denmark
| |
Collapse
|
39
|
Mu M, Gao P, Yang Q, He J, Wu F, Han X, Guo S, Qian Z, Song C. Alveolar Epithelial Cells Promote IGF-1 Production by Alveolar Macrophages Through TGF-β to Suppress Endogenous Inflammatory Signals. Front Immunol 2020; 11:1585. [PMID: 32793225 PMCID: PMC7385185 DOI: 10.3389/fimmu.2020.01585] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Accepted: 06/15/2020] [Indexed: 12/22/2022] Open
Abstract
To maintain alveolar gas exchange, the alveolar surface has to limit unnecessary inflammatory responses. This involves crosstalk between alveolar epithelial cells (AECs) and alveolar macrophages (AMs) in response to damaging factors. We recently showed that insulin-like growth factor (IGF)-1 regulates the phagocytosis of AECs. AMs secrete IGF-1 into the bronchoalveolar lavage fluid (BALF) in response to inflammatory stimuli. However, whether AECs regulate the production of IGF-1 by AMs in response to inflammatory signals remains unclear, as well as the role of IGF-1 in controlling the alveolar balance in the crosstalk between AMs and AECs under inflammatory conditions. In this study, we demonstrated that IGF-1 was upregulated in BALF and lung tissues of acute lung injury (ALI) mice, and that the increased IGF-1 was mainly derived from AMs. In vitro experiments showed that the production and secretion of IGF-1 by AMs as well as the expression of TGF-β were increased in LPS-stimulated AEC-conditioned medium (AEC-CM). Pharmacological blocking of TGF-β in AECs and addition of TGF-β neutralizing antibody to AEC-CM suggested that this AEC-derived cytokine mediates the increased production and secretion of IGF-1 from AMs. Blocking TGF-β synthesis or treatment with TGF-β neutralizing antibody attenuated the increase of IGF-1 in BALF in ALI mice. TGF-β induced the production of IGF-1 by AMs through the PI3K/Akt signaling pathway. IGF-1 prevented LPS-induced p38 MAPK activation and the expression of the inflammatory factors MCP-1, TNF-α, and IL-1β in AECs. However, IGF-1 upregulated PPARγ to increase the phagocytosis of apoptotic cells by AECs. Intratracheal instillation of IGF-1 decreased the number of polymorphonuclear neutrophils in BALF of ALI model mice, reduced alveolar congestion and edema, and suppressed inflammatory cell infiltration in lung tissues. These results elucidated a mechanism by which AECs used TGF-β to regulate IGF-1 production from AMs to attenuate endogenous inflammatory signals during alveolar inflammation.
Collapse
Affiliation(s)
- Mimi Mu
- Department of Immunology, School of Laboratory Medicine, Bengbu Medical College, Bengbu, China.,Anhui Provincial Key Laboratory of Infection and Immunity, Bengbu Medical College, Bengbu, China.,Anhui Province Key Laboratory of Immunology in Chronic Diseases, Bengbu Medical College, Bengbu, China
| | - Peiyu Gao
- Department of Immunology, School of Laboratory Medicine, Bengbu Medical College, Bengbu, China.,Anhui Provincial Key Laboratory of Infection and Immunity, Bengbu Medical College, Bengbu, China.,Anhui Province Key Laboratory of Immunology in Chronic Diseases, Bengbu Medical College, Bengbu, China
| | - Qian Yang
- Department of Immunology, School of Laboratory Medicine, Bengbu Medical College, Bengbu, China.,Anhui Provincial Key Laboratory of Infection and Immunity, Bengbu Medical College, Bengbu, China.,Anhui Province Key Laboratory of Immunology in Chronic Diseases, Bengbu Medical College, Bengbu, China
| | - Jing He
- Department of Immunology, School of Laboratory Medicine, Bengbu Medical College, Bengbu, China.,Anhui Provincial Key Laboratory of Infection and Immunity, Bengbu Medical College, Bengbu, China.,Anhui Province Key Laboratory of Immunology in Chronic Diseases, Bengbu Medical College, Bengbu, China
| | - Fengjiao Wu
- Department of Immunology, School of Laboratory Medicine, Bengbu Medical College, Bengbu, China.,Anhui Provincial Key Laboratory of Infection and Immunity, Bengbu Medical College, Bengbu, China.,Anhui Province Key Laboratory of Immunology in Chronic Diseases, Bengbu Medical College, Bengbu, China
| | - Xue Han
- Department of Immunology, School of Laboratory Medicine, Bengbu Medical College, Bengbu, China.,Anhui Provincial Key Laboratory of Infection and Immunity, Bengbu Medical College, Bengbu, China.,Anhui Province Key Laboratory of Immunology in Chronic Diseases, Bengbu Medical College, Bengbu, China
| | - Shujun Guo
- Department of Immunology, School of Laboratory Medicine, Bengbu Medical College, Bengbu, China.,Anhui Provincial Key Laboratory of Infection and Immunity, Bengbu Medical College, Bengbu, China.,Anhui Province Key Laboratory of Immunology in Chronic Diseases, Bengbu Medical College, Bengbu, China
| | - Zhongqing Qian
- Department of Immunology, School of Laboratory Medicine, Bengbu Medical College, Bengbu, China.,Anhui Provincial Key Laboratory of Infection and Immunity, Bengbu Medical College, Bengbu, China.,Anhui Province Key Laboratory of Immunology in Chronic Diseases, Bengbu Medical College, Bengbu, China
| | - Chuanwang Song
- Department of Immunology, School of Laboratory Medicine, Bengbu Medical College, Bengbu, China.,Anhui Provincial Key Laboratory of Infection and Immunity, Bengbu Medical College, Bengbu, China.,Anhui Province Key Laboratory of Immunology in Chronic Diseases, Bengbu Medical College, Bengbu, China
| |
Collapse
|
40
|
Evaluation of the retinal layers and microvasculature in patients with acromegaly: a case-control OCT angiography study. Eye (Lond) 2020; 35:523-527. [PMID: 32346112 DOI: 10.1038/s41433-020-0884-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2019] [Revised: 03/31/2020] [Accepted: 04/08/2020] [Indexed: 11/08/2022] Open
Abstract
BACKGROUND The aim of study was to evaluate the retinal layers and macular capillary network with OCTA in acromegaly patients, to compare with healthy population. METHODS In this prospective, observational, and comparative study, 40 acromegaly patients and 40 healthy control participants were included. Serum IGF-1 levels and disease duration of all patients were noted. Macular layers and angiography scanning was performed with a Zeiss Cirrus 5000 OCTA system. Macular thickness, RNFL, and GC-IPL values were obtained. For central vessel and perfusion density, central 6 mm was obtained and was evaluated by dividing into three groups (inner, outer, full). FAZ parameters were evaluated dividing into three groups (area, perimeter, circularity index). Analysis of the data was performed with the SPSS for Windows. RESULTS There was no significant difference between the patient group and the control group in terms of age, gender, best corrected visual acuity (BCVA), spherical equivalent (SE), intraocular pressure (IOP), and axial length (AL). The mean follow-up period after diagnosis was 11.0 ± 5.5 years. Central and mean macular thicknesses were also significantly higher in the acromegaly group (p < 0.05). Superior, inferior, and average RNFL thicknesses were also significantly thinner in the acromegaly group (p < 0.05). When OCTA parameters were compared between groups, there was a significant decrease in central vessel density (CVD) and central perfusion density (CPD) values in all regions in acromegaly group compared with controls (p < 0.05). CONCLUSION Our findings with OCTA show that acromegaly causes a significant capillary network decrease according to the healthy subjects.
Collapse
|
41
|
Li Y, Cheng T, Wan C, Cang Y. circRNA_0084043 contributes to the progression of diabetic retinopathy via sponging miR-140-3p and inducing TGFA gene expression in retinal pigment epithelial cells. Gene 2020; 747:144653. [PMID: 32259630 DOI: 10.1016/j.gene.2020.144653] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2020] [Revised: 03/20/2020] [Accepted: 04/03/2020] [Indexed: 01/22/2023]
Abstract
Diabetic retinopathy (DR) is a frequent complication of diabetes and it can lead to visual impairment and blindness. However, the mechanism of their regulation remains little known. circRNAs can function as crucial competing endogenous RNA, which can sponge corresponding miRNAs and affect mRNA expression in various diseases, including DR. In our current research, we observed that circRNA_0084043 was elevated in high glucose (HG)-incubated ARPE-19 cells. Then, we focused on whether and how circRNA_0084043 participated in retinal vascular dysfunction under conditions diabetes. Apoptosis, inflammation and oxidative stress are hallmark of DR progression. This work was aimed to investigate the signaling mechanisms of circRNA_0084043 in these pathogenesis of DR. We discovered loss of circRNA_0084043 significantly increased cell survival and repressed HG-triggered apoptosis. In addition, knockdown of circRNA_0084043 remarkably reduced oxidative stress as evidenced by the down-regulated malondialdehyde (MDA) content, enhanced activities of Super Oxide Dismutase (SOD) and Glutathione peroxidase (GSH-PX). Addition, silence of circRNA_0084043 effectively restrained HG-stimulated inflammation as proved by repressing inflammatory cytokines Tumor Necrosis Factor α (TNF-α), Interleukin 6 (IL-6) and Cox-2 in ARPE-19 cells. Subsequently, we successfully predicted that miR-140-3p was a downstream target of circRNA_0084043, which could be negatively regulated by circRNA_0084043. Mechanistically, loss of miR-140-3p abrogated the beneficial effects of circRNA_0084043 siRNA on ARPE-19 cells. Transforming Growth Factor alpha (TGFA) can exhibit a role in multiple diseases. Taken these together, these data demonstrated that loss of circRNA_0084043 depressed HG-induced damage via sponging miR-140-3p and regulating TGFA.
Collapse
Affiliation(s)
- Ying Li
- Physical Examination Center, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Ting Cheng
- General Department of Houhu, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430014, China
| | - Chengliang Wan
- Department of General Surgery, Kunming Children's Hospital, Kunming, China
| | - Yanhong Cang
- Nursing Department, The Affiliated Huai'an Hospital of Xuzhou Medical University and The Second People's Hospital of Huai'an, 62 Huaihai Road (S.), Huai'an 223002, China.
| |
Collapse
|
42
|
Jiang S, Kametani M, Chen DF. Adaptive Immunity: New Aspects of Pathogenesis Underlying Neurodegeneration in Glaucoma and Optic Neuropathy. Front Immunol 2020; 11:65. [PMID: 32117239 PMCID: PMC7031201 DOI: 10.3389/fimmu.2020.00065] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2019] [Accepted: 01/10/2020] [Indexed: 01/04/2023] Open
Abstract
Glaucoma is a globally unmet medical challenge and the most prevalent neurodegenerative disease, which permanently damages the optic nerve and retinal ganglion cells (RGCs), leading to irreversible blindness. Present therapies target solely at lowering intraocular ocular pressure (IOP), a major risk factor of the disease; however, elevated IOP is neither necessary nor sufficient to cause glaucoma. Glaucomatous RGC and nerve fiber loss also occur in individuals with normal IOP. Recent studies have provided evidence indicating a link between elevated IOP and T cell-mediated autoimmune responses, particularly that are specific to heat shock proteins (HSPs), underlying the pathogenesis of neurodegeneration in glaucoma. Reactive glial responses and low-grade inflammation may initially represent an adaptive reaction of the retina to primary stress stimuli; whereas, sustained and excessive glial reactions lead to expanded immune responses, including adaptive immunity, that contribute to progressive neural damage in glaucoma. Emerging data suggest a similar mechanism in play in causing neurodegeneration of other forms of optic neuropathy, such as that resulted from acute ischemia and traumatic injuries. These studies may lead to the paradigm shift and offer a new basis for the development of novel mechanism-based diagnosis, therapy, and preventive interventions for glaucoma. As HSPs are induced under various conditions of neural stress and damage in the brain and spinal cord, these findings may have broader implications for our elucidating of the etiology of other neurodegenerative disorders in the central nervous system.
Collapse
Affiliation(s)
- Shuhong Jiang
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA, United States
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Marie Kametani
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA, United States
| | - Dong Feng Chen
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA, United States
| |
Collapse
|
43
|
Lu BW, Xie LK. Potential applications of artemisinins in ocular diseases. Int J Ophthalmol 2019; 12:1793-1800. [PMID: 31741871 DOI: 10.18240/ijo.2019.11.20] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2018] [Accepted: 04/17/2019] [Indexed: 02/07/2023] Open
Abstract
Artemisinin, also named qinghaosu, is a family of sesquiterpene trioxane lactone originally derived from the sweet wormwood plant (Artemisia annua), which is a traditional Chinese herb that has been universally used as anti-malarial agents for many years. Evidence has accumulated during the past few years which demonstrated the protective effects of artemisinin and its derivatives (artemisinins) in several other diseases beyond malaria, including cancers, autoimmune disorders, inflammatory diseases, viral and other parasite-related infections. Recently, this long-considered anti-malarial agent has been proved to possess anti-oxidant, anti-inflammatory, anti-apoptotic and anti-excitotoxic properties, which make it a potential treatment option for the ocular environment. In this review, we first described the overview of artemisinins, highlighting the activity of artemisinins to other diseases beyond malaria and the mechanisms of these actions. We then emphasized the main points of published results of using artemisinins in targeting ocular disorders, including uveitis, retinoblastoma, retinal neurodegenerative diseases and ocular neovascularization. To conclude, we believe that artemisinins could also be used as a promising therapeutic drug for ocular diseases, especially retinal vascular diseases in the near future.
Collapse
Affiliation(s)
- Bing-Wen Lu
- Department of Ophthalmology, Eye Hospital, China Academy of Chinese Medical Sciences, Beijing 100400, China
| | - Li-Ke Xie
- Department of Ophthalmology, Eye Hospital, China Academy of Chinese Medical Sciences, Beijing 100400, China
| |
Collapse
|
44
|
Litwińska Z, Sobuś A, Łuczkowska K, Grabowicz A, Mozolewska-Piotrowska K, Safranow K, Kawa MP, Machaliński B, Machalińska A. The Interplay Between Systemic Inflammatory Factors and MicroRNAs in Age-Related Macular Degeneration. Front Aging Neurosci 2019; 11:286. [PMID: 31695606 PMCID: PMC6817913 DOI: 10.3389/fnagi.2019.00286] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2019] [Accepted: 10/04/2019] [Indexed: 12/20/2022] Open
Abstract
We aimed to explore the expression of systemic inflammatory factors and selected intracellular miRNAs that regulate inflammatory signaling pathways potentially involved in age-related macular degeneration (AMD) pathogenesis. A total of 179 patients with wet AMD, 175 with dry AMD and 121 controls were enrolled in the study. Soluble inflammatory factors were analyzed in plasma samples using Luminex technology. Expression of selected miRNAs was analyzed in isolated nucleated peripheral blood cells (PBNCs) using real-time qPCR. Wet AMD was an independent factor associated with higher concentrations of IL-6 (β = +0.24, p = 0.0004), GM-CSF (β = +0.31, p < 0.001), IFN-γ (β = +0.58, p < 0.001), higher expression of miRNA-23a-3p (β = +0.60, p < 0.0001), miRNA-30b (β = +0.32, p < 0.0001), miRNA-191-5p (β = +0.28, p < 0.0001) and lower concentration of IL-1β (β = −0.25, p = 0.0003), IL-5 (β = −0.45, p < 0.001), IL-10 (β = −0.45, p < 0.001), IL-12 (β = −0.35, p < 0.001), lower expression of miRNA-16-5p (β = −0.31, p < 0.0001), miRNA-17-3p (β = −0.18, p = 0.01), miRNA-150-5p (β = −0.18, p = 0.01) and miRNA-155-5p (β = −0.47, p < 0.0001). Multivariate analysis revealed that dry AMD was an independent factor associated with higher concentration of GM-CSF (β = +0.34, p < 0.001), IL-6 (β = +0.13, p = 0.05), higher expression of miRNA-23a-3p (β = +0.60, p < 0.0001), miRNA-126-3p (β = +0.23, p = 0.0005), miRNA-126-5p (β = +0.16, p = 0.01), miRNA 146a (β = +0.14, p = 0.03), and mRNA191-5p (β = +0.15, p = 0.03) and lower concentrations of TNF-α (β = +0.24, p = 0.0004), IL-1β (β = −0.39, p < 0.001), IL-2 (β = −0.20, p = 0.003), IL-5 (β = −0.54, p < 0.001), IL-10 (β = −0.56, p < 0.001), IL-12 (β = −0.51, p < 0.001), lower expression of miRNA-16-5p (β = −0.23, p = 0.0004), miRNA-17-3p (β = −0.20, p = 0.003) and miRNA-17-5p (β = −0.19, p = 0.004). Negative correlations between visual acuity and WBC, lymphocyte count, TNF-α, IL-1 β, IL-2, IL-4, IL-6, IL-10 concentrations and miRNA-191-5p, as well as positive correlations between visual acuity and miRNA-126-3p, -126-5p, and -155-5p PBNCs expression were found in AMD patients. No such correlations were found in the control group. Our results may suggest the role of both intra- and extracellular mechanisms implicated in inflammatory response regulation in multifactorial AMD pathogenesis.
Collapse
Affiliation(s)
- Zofia Litwińska
- Department of General Pathology, Pomeranian Medical University, Szczecin, Poland
| | - Anna Sobuś
- Department of General Pathology, Pomeranian Medical University, Szczecin, Poland
| | - Karolina Łuczkowska
- Department of General Pathology, Pomeranian Medical University, Szczecin, Poland
| | - Aleksandra Grabowicz
- First Department of Ophthalmology, Pomeranian Medical University, Szczecin, Poland
| | | | - Krzysztof Safranow
- Department of Biochemistry and Medical Chemistry, Pomeranian Medical University, Szczecin, Poland
| | - Miłosz Piotr Kawa
- Department of General Pathology, Pomeranian Medical University, Szczecin, Poland
| | - Bogusław Machaliński
- Department of General Pathology, Pomeranian Medical University, Szczecin, Poland
| | - Anna Machalińska
- First Department of Ophthalmology, Pomeranian Medical University, Szczecin, Poland
| |
Collapse
|
45
|
Myhre CL, Thygesen C, Villadsen B, Vollerup J, Ilkjær L, Krohn KT, Grebing M, Zhao S, Khan AM, Dissing-Olesen L, Jensen MS, Babcock AA, Finsen B. Microglia Express Insulin-Like Growth Factor-1 in the Hippocampus of Aged APP swe/PS1 ΔE9 Transgenic Mice. Front Cell Neurosci 2019; 13:308. [PMID: 31417357 PMCID: PMC6682662 DOI: 10.3389/fncel.2019.00308] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2018] [Accepted: 06/24/2019] [Indexed: 11/14/2022] Open
Abstract
Insulin-like growth factor-1 (IGF-1) is a pleiotropic molecule with neurotrophic and immunomodulatory functions. Knowing the capacity of chronically activated microglia to produce IGF-1 may therefore show essential to promote beneficial microglial functions in Alzheimer's disease (AD). Here, we investigated the expression of IGF-1 mRNA and IGF-1 along with the expression of tumor necrosis factor (TNF) mRNA, and the amyloid-β (Aβ) plaque load in the hippocampus of 3- to 24-month-old APPswe/PS1ΔE9 transgenic (Tg) and wild-type (WT) mice. As IGF-1, in particular, is implicated in neurogenesis we also monitored the proliferation of cells in the subgranular zone (sgz) of the dentate gyrus. We found that the Aβ plaque load reached its maximum in aged 21- and 24-month-old APPswe/PS1ΔE9 Tg mice, and that microglial reactivity and hippocampal IGF-1 and TNF mRNA levels were significantly elevated in aged APPswe/PS1ΔE9 Tg mice. The sgz cell proliferation decreased with age, regardless of genotype and increased IGF-1/TNF mRNA levels. Interestingly, IGF-1 mRNA was expressed in subsets of sgz cells, likely neuroblasts, and neurons in both genotypes, regardless of age, as well as in glial-like cells. By double in situ hybridization these were shown to be IGF1 mRNA+ CD11b mRNA+ cells, i.e., IGF-1 mRNA-expressing microglia. Quantification showed a 2-fold increase in the number of microglia and IGF-1 mRNA-expressing microglia in the molecular layer of the dentate gyrus in aged APPswe/PS1ΔE9 Tg mice. Double-immunofluorescence showed that IGF-1 was expressed in a subset of Aβ plaque-associated CD11b+ microglia and in several subsets of neurons. Exposure of primary murine microglia and BV2 cells to Aβ42 did not affect IGF-1 mRNA expression. IGF-1 mRNA levels remained constant in WT mice with aging, unlike TNF mRNA levels which increased with aging. In conclusion, our results suggest that the increased IGF-1 mRNA levels can be ascribed to a larger number of IGF-1 mRNA-expressing microglia in the aged APPswe/PS1ΔE9 Tg mice. The finding that subsets of microglia retain the capacity to express IGF-1 mRNA and IGF-1 in the aged APPswe/PS1ΔE9 Tg mice is encouraging, considering the beneficial therapeutic potential of modulating microglial production of IGF-1 in AD.
Collapse
Affiliation(s)
- Christa Løth Myhre
- Department of Neurobiology, Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | - Camilla Thygesen
- Department of Neurobiology, Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark
- Brain Research – Inter-Disciplinary Guided Excellence, Department of Clinical Research, University of Southern Denmark, Odense, Denmark
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, Odense, Denmark
| | - Birgitte Villadsen
- Department of Neurobiology, Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark
- Brain Research – Inter-Disciplinary Guided Excellence, Department of Clinical Research, University of Southern Denmark, Odense, Denmark
| | - Jeanette Vollerup
- Department of Neurobiology, Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark
- Brain Research – Inter-Disciplinary Guided Excellence, Department of Clinical Research, University of Southern Denmark, Odense, Denmark
| | - Laura Ilkjær
- Department of Neurobiology, Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | - Katrine Tækker Krohn
- Department of Neurobiology, Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark
- Brain Research – Inter-Disciplinary Guided Excellence, Department of Clinical Research, University of Southern Denmark, Odense, Denmark
| | - Manuela Grebing
- Department of Neurobiology, Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | - Shuainan Zhao
- Department of Neurobiology, Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark
- Brain Research – Inter-Disciplinary Guided Excellence, Department of Clinical Research, University of Southern Denmark, Odense, Denmark
| | - Asif Manzoor Khan
- Department of Neurobiology, Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | - Lasse Dissing-Olesen
- Department of Neurobiology, Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | | | - Alicia A. Babcock
- Department of Neurobiology, Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | - Bente Finsen
- Department of Neurobiology, Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark
- Brain Research – Inter-Disciplinary Guided Excellence, Department of Clinical Research, University of Southern Denmark, Odense, Denmark
| |
Collapse
|
46
|
McGinn TE, Galicia CA, Leoni DC, Partington N, Mitchell DM, Stenkamp DL. Rewiring the Regenerated Zebrafish Retina: Reemergence of Bipolar Neurons and Cone-Bipolar Circuitry Following an Inner Retinal Lesion. Front Cell Dev Biol 2019; 7:95. [PMID: 31245369 PMCID: PMC6562337 DOI: 10.3389/fcell.2019.00095] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2018] [Accepted: 05/17/2019] [Indexed: 11/13/2022] Open
Abstract
We previously reported strikingly normal morphologies and functional connectivities of regenerated retinal bipolar neurons (BPs) in zebrafish retinas sampled 60 days after a ouabain-mediated lesion of inner retinal neurons (60 DPI) (McGinn et al., 2018). Here we report early steps in the birth of BPs and formation of their dendritic trees and axonal arbors during regeneration. Adult zebrafish were subjected to ouabain-mediated lesion that destroys inner retinal neurons but spares photoreceptors and Müller glia, and were sampled at 13, 17, and 21 DPI, a timeframe over which plexiform layers reemerge. We show that this timeframe corresponds to reemergence of two populations of BPs (PKCα+ and nyx::mYFP+). Sequential BrdU, EdU incorporation reveals that similar fractions of PKCα+ BPs and HuC/D+ amacrine/ganglion cells are regenerated concurrently, suggesting that the sequence of neuronal production during retinal regeneration does not strictly match that observed during embryonic development. Further, accumulation of regenerated BPs appears protracted, at least through 21 DPI. The existence of isolated, nyx::mYFP+ BPs allowed examination of cytological detail through confocal microscopy, image tracing, morphometric analyses, identification of cone synaptic contacts, and rendering/visualization. Apically-projecting neurites (=dendrites) of regenerated BPs sampled at 13, 17, and 21 DPI are either truncated, or display smaller dendritic trees when compared to controls. In cases where BP dendrites reach the outer plexiform layer (OPL), numbers of dendritic tips are similar to those of controls at all sampling times. Further, by 13-17 DPI, BPs with dendritic tips reaching the outer nuclear layer (ONL) show patterns of photoreceptor connections that are statistically indistinguishable from controls, while those sampled at 21 DPI slightly favor contacts with double cone synaptic terminals over those of blue-sensitive cones. These findings suggest that once regenerated BP dendrites reach the OPL, normal photoreceptor connectomes are established, albeit with some plasticity. Through 17 DPI, some basally-projecting neurites (=axons) of regenerated nyx::mYFP+ BPs traverse long distances, branch into inappropriate layers, or appear to abruptly terminate. These findings suggest that, after a tissue-disrupting lesion, regeneration of inner retinal neurons is a dynamic process that includes ongoing genesis of new neurons and changes in BP morphology.
Collapse
Affiliation(s)
- Timothy E McGinn
- Department of Biological Sciences, University of Idaho, Moscow, ID, United States
| | - Carlos A Galicia
- Department of Biological Sciences, University of Idaho, Moscow, ID, United States
| | - Dylan C Leoni
- Department of Biological Sciences, University of Idaho, Moscow, ID, United States
| | - Natalie Partington
- Department of Biology, Brigham Young University-Idaho, Rexburg, ID, United States
| | - Diana M Mitchell
- Department of Biological Sciences, University of Idaho, Moscow, ID, United States
| | - Deborah L Stenkamp
- Department of Biological Sciences, University of Idaho, Moscow, ID, United States
| |
Collapse
|
47
|
Mao XB, Cheng YH, Xu YY. miR-204-5p promotes diabetic retinopathy development via downregulation of microtubule-associated protein 1 light chain 3. Exp Ther Med 2019; 17:2945-2952. [PMID: 30936964 PMCID: PMC6434256 DOI: 10.3892/etm.2019.7327] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2017] [Accepted: 12/12/2018] [Indexed: 12/14/2022] Open
Abstract
Diabetic retinopathy (DR) is a chronic and progressive complication of diabetes mellitus. DR impairs sight due to neuronal and vascular dysfunction in the retina. It is critical to investigate the pathogenesis of DR to develop effective treatment. In the present study, a streptozotocin (STZ)-induced diabetic rat model was constructed and the expression of microRNA (miR)-204-5p and vascular endothelial growth factor (VEGF) were determined. Immunohistochemistry, reverse transcription-quantitative polymerase chain reaction (RT-qPCR) and western blotting were employed to detect the effects of miR-204-5p on the expression of microtubule-associated protein 1 light chain 3 (LC3B). RT-qPCR analysis demonstrated that miR-204-5p and VEGF were significantly upregulated in the retina tissue of diabetic rats compared with the control group (P<0.01). Immunohistochemistry and western blotting revealed that the protein expression levels of LC3B-II and the ratio of LC3B-II/LC3B-I were significantly suppressed in the diabetes group compared with the control (P<0.01). In retinal tissues, anti-miR-204-5p treatment significantly enhanced the protein expression levels of LC3B-II and the ratio of LC3B-II/LC3B-I and these levels were significantly reduced in response to miR-204-5p mimic treatment compared with the negative miR control (P<0.01). In rat retinal endothelial cells isolated from diabetic rats, anti-miR-204-5p treatment increased the number of autophagic vacuoles, and significantly promoted LC3B-II expression and the LC3B-II/LC3B-I ratio compared with the negative control (P<0.01). The results of the present study revealed that miR-204-5p downregulated the expression of LC3B-II to inhibit autophagy in DR. Therefore, miR-204-5p may be considered as a novel effective therapeutic target during the development of DR.
Collapse
Affiliation(s)
- Xin-Bang Mao
- Department of Ophthalmology, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Yan-Hua Cheng
- Department of Ophthalmology, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Yan-Ying Xu
- Department of Ophthalmology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310016, P.R. China
| |
Collapse
|
48
|
Fuentes-Santamaría V, Alvarado JC, Rodríguez-de la Rosa L, Juiz JM, Varela-Nieto I. Neuroglial Involvement in Abnormal Glutamate Transport in the Cochlear Nuclei of the Igf1 -/- Mouse. Front Cell Neurosci 2019; 13:67. [PMID: 30881288 PMCID: PMC6405628 DOI: 10.3389/fncel.2019.00067] [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: 11/08/2018] [Accepted: 02/11/2019] [Indexed: 12/25/2022] Open
Abstract
Insulin-like growth factor 1 (IGF-1) is a powerful regulator of synaptic activity and a deficit in this protein has a profound impact on neurotransmission, mostly on excitatory synapses in both the developing and mature auditory system. Adult Igf1−/− mice are animal models for the study of human syndromic deafness; they show altered cochlear projection patterns into abnormally developed auditory neurons along with impaired glutamate uptake in the cochlear nuclei, phenomena that probably reflect disruptions in neuronal circuits. To determine the cellular mechanisms that might be involved in regulating excitatory synaptic plasticity in 4-month-old Igf1−/− mice, modifications to neuroglia, astroglial glutamate transporters (GLTs) and metabotropic glutamate receptors (mGluRs) were assessed in the cochlear nuclei. The Igf1−/− mice show significant decreases in IBA1 (an ionized calcium-binding adapter) and glial fibrillary acidic protein (GFAP) mRNA expression and protein accumulation, as well as dampened mGluR expression in conjunction with enhanced glutamate transporter 1 (GLT1) expression. By contrast, no differences were observed in the expression of glutamate aspartate transporter (GLAST) between these Igf1−/− mice and their heterozygous or wildtype littermates. These observations suggest that congenital IGF-1 deficiency may lead to alterations in microglia and astrocytes, an upregulation of GLT1, and the downregulation of groups I, II and III mGluRs. Understanding the molecular, biochemical and morphological mechanisms underlying neuronal plasticity in a mouse model of hearing deficits will give us insight into new therapeutic strategies that could help to maintain or even improve residual hearing when human deafness is related to IGF-1 deficiency.
Collapse
Affiliation(s)
- Veronica Fuentes-Santamaría
- Instituto de Investigación en Discapacidades Neurológicas (IDINE), Facultad de Medicina, Universidad de Castilla-La Mancha, Albacete, Spain
| | - Juan C Alvarado
- Instituto de Investigación en Discapacidades Neurológicas (IDINE), Facultad de Medicina, Universidad de Castilla-La Mancha, Albacete, Spain
| | - Lourdes Rodríguez-de la Rosa
- Grupo de Neurobiología de la Audición, Instituto de Investigaciones Biomédicas Alberto Sols, Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid, Madrid, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), CIBER MP, Instituto de Salud Carlos III, Madrid, Spain.,Instituto de Investigación Sanitaria del Hospital Universitario La Paz (IdiPAZ), Madrid, Spain
| | - José M Juiz
- Instituto de Investigación en Discapacidades Neurológicas (IDINE), Facultad de Medicina, Universidad de Castilla-La Mancha, Albacete, Spain
| | - Isabel Varela-Nieto
- Grupo de Neurobiología de la Audición, Instituto de Investigaciones Biomédicas Alberto Sols, Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid, Madrid, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), CIBER MP, Instituto de Salud Carlos III, Madrid, Spain.,Instituto de Investigación Sanitaria del Hospital Universitario La Paz (IdiPAZ), Madrid, Spain
| |
Collapse
|
49
|
Kramer J, Chirco KR, Lamba DA. Immunological Considerations for Retinal Stem Cell Therapy. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1186:99-119. [PMID: 31654387 DOI: 10.1007/978-3-030-28471-8_4] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
There is an increasing effort toward generating replacement cells for neuronal application due to the nonregenerative nature of these tissues. While much progress has been made toward developing methodologies to generate these cells, there have been limited improvements in functional restoration. Some of these are linked to the degenerative and often nonreceptive microenvironment that the new cells need to integrate into. In this chapter, we will focus on the status and role of the immune microenvironment of the retina during homeostasis and disease states. We will review changes in both innate and adaptive immunity as well as the role of immune rejection in stem cell replacement therapies. The chapter will end with a discussion of immune-modulatory strategies that have helped to ameliorate these effects and could potentially improve functional outcome for cell replacement therapies for the eye.
Collapse
Affiliation(s)
- Joshua Kramer
- Buck Institute for Research on Aging, Novato, CA, USA
| | | | - Deepak A Lamba
- Department of Ophthalmology, University of California San Francisco, San Francisco, CA, USA. .,Buck Institute for Research on Aging, Novato, CA, USA.
| |
Collapse
|