1
|
Lu T, Shang J, Pu S, Xu Y, Sun X, Gao X. The role of microglia in the development of diabetic retinopathy and its potential clinical application. Hum Cell 2025; 38:101. [PMID: 40392429 DOI: 10.1007/s13577-025-01226-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2024] [Accepted: 04/21/2025] [Indexed: 05/22/2025]
Abstract
Lately, research on the function of microglia in diabetic retinopathy (DR) is becoming increasingly focused. Microglia are immune cells that dwell in the central nervous system and are crucial to the pathophysiology of DR. According to studies, a hyperglycemic environment can activate microglia, bringing them out of a resting state to an active state. This allows them to release a variety of inflammatory factors and chemokines, which can then cause retinal inflammatory reactions. When it comes to angiogenesis in DR, activated microglia release a variety of angiogenic substances, such as vascular endothelial growth factor (VEGF), to create aberrant new blood vessels. Moreover, microglia contribute to the retina's oxidative stress process by generating and releasing reactive oxygen and nitrogen-free radicals, which exacerbates retinal damage. Researchers have proposed a variety of strategies for the activation of microglia and the inflammatory response it triggers. By inhibiting the excessive activation of microglia and reducing the release of inflammatory factors, the inflammatory response and damage to the retina can be alleviated. Drugs that interfere with retinal microglia can also be used to regulate vascular damage and inhibit the formation of new blood vessels. In addition, antioxidants are used to remove reactive oxygen and free radicals, reduce oxidative stress levels, and protect retinal cells. These therapeutic strategies aim to achieve the purpose of treating DR by regulating the function of microglia. Thus, we highlight the possibility that therapy aimed at microglia could offer fresh ideas for treating DR.
Collapse
Affiliation(s)
- Tingting Lu
- The First Affiliated Hospital of Harbin Medical University, Harbin, People's Republic of China
| | - Jiameng Shang
- The First Affiliated Hospital of Harbin Medical University, Harbin, People's Republic of China
| | - Shengdan Pu
- The First Affiliated Hospital of Harbin Medical University, Harbin, People's Republic of China
| | - Yuxin Xu
- The First Affiliated Hospital of Harbin Medical University, Harbin, People's Republic of China
| | - Xiaotong Sun
- The First Affiliated Hospital of Harbin Medical University, Harbin, People's Republic of China
| | - Xinyuan Gao
- The First Affiliated Hospital of Harbin Medical University, Harbin, People's Republic of China.
| |
Collapse
|
2
|
Harley O, Amelia YS, Gustianty E, Soetedjo NNM, Kartasasmita AS. Exploring leukocyte differential count ratio profiles as inflammatory biomarkers in diabetic retinopathy: a systematic review and meta-analysis. BMC Ophthalmol 2025; 25:265. [PMID: 40312699 PMCID: PMC12044949 DOI: 10.1186/s12886-025-04075-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2024] [Accepted: 04/16/2025] [Indexed: 05/03/2025] Open
Abstract
BACKGROUND Diabetic retinopathy (DR) is increasingly prevalent and a major cause of irreversible blindness, particularly in developing countries. Limited access to ophthalmologists often leads to delayed diagnosis, emphasizing the need for more affordable and widely accessible screening methods to facilitate early identification. Recently, several studies have demonstrated variability in findings regarding the relationship between leukocyte differential count ratio biomarkers and DR. This study aims to investigate the association between leukocyte differential count ratios-NLR (Neutrophil-to-Lymphocyte Ratio), PLR (Platelet-to-Lymphocyte Ratio), MLR (Monocyte-to-Lymphocyte Ratio), and SII (Systemic Immune-Inflammation Index)-and the stages of diabetic retinopathy (DR). METHODS A comprehensive literature search was conducted across several databases up to September 2024, with a focus on identifying studies examining the relationship between the leukocyte differential count ratio profiles and diabetic retinopathy. Review Manager was used to conduct the meta-analyses. The Newcastle Ottawa Scale (NOS) were used to assess the included studies. RESULTS A total of 38 studies were included in the systematic review and 27 studies were included in the meta-analysis. The mean differences in the NLR and PLR values were significantly different among the groups and were higher in the PDR group (0.68 (95%CI 0.42-0.95, p < 0.05) and 19.57 (95%CI 10.68-28.46, p < 0.05; respectively). These findings were followed by significant differences in SII value 202.53 (95% CI 196.19-208.86, p < 0.05). Moreover, the MLR values were not significantly different among the groups (p > 0.05). CONCLUSION NLR, PLR, and SII are associated with both the presence and progression of DR, with increasing levels of NLR and PLR reflecting a higher risk and severity of the disease. However, it is still necessary to justify the need to combine them with other clinical parameters to confirm the diagnosis.
Collapse
Affiliation(s)
- Ohisa Harley
- Doctoral Program in Medical Sciences, Faculty of Medicine, Padjadjaran University, Terusan Ekologi- No 7, Ir. Soekarno Street Km 21, Jatinangor, Bandung, West Java, 45363, Indonesia.
- Netra Eye Clinic, Sumatera Street No. 46 - 68, Bandung, West Java, 40114, Indonesia.
| | - Yufilia Suci Amelia
- Netra Eye Clinic, Sumatera Street No. 46 - 68, Bandung, West Java, 40114, Indonesia
| | - Elsa Gustianty
- Netra Eye Clinic, Sumatera Street No. 46 - 68, Bandung, West Java, 40114, Indonesia
- Departement of Ophthalmology, Faculty of Medicine, Padjadjaran University, Ir. Soekarno Street Km 21, Bandung, West Java, 45363, Indonesia
- Cicendo Hospital National Eye Center, Cicendo Street No.4, Babakan Ciamis, Bandung, West Java, 40117, Indonesia
| | - Nanny N M Soetedjo
- Departement of Endocrinology and Internal Medicine, Faculty of Medicine, Padjadjaran University, Ir. Soekarno Street Km 21, Jatinangor, Bandung, West Java, 45363, Indonesia
| | - Arief S Kartasasmita
- Netra Eye Clinic, Sumatera Street No. 46 - 68, Bandung, West Java, 40114, Indonesia
- Departement of Ophthalmology, Faculty of Medicine, Padjadjaran University, Ir. Soekarno Street Km 21, Bandung, West Java, 45363, Indonesia
- Cicendo Hospital National Eye Center, Cicendo Street No.4, Babakan Ciamis, Bandung, West Java, 40117, Indonesia
| |
Collapse
|
3
|
Murugesan S, Addis DR, Hussey H, Powell MF, Saravanakumar L, Sturdivant AB, Sinkey RG, Tubinis MD, Massey ZR, Patton C, Mobley JA, Tita AN, Jilling T, Berkowitz DE. Decreased Extracellular Vesicle Vasorin in Severe Preeclampsia Plasma Mediates Endothelial Dysfunction. J Am Heart Assoc 2025; 14:e037242. [PMID: 40118804 DOI: 10.1161/jaha.124.037242] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Accepted: 01/30/2025] [Indexed: 03/23/2025]
Abstract
BACKGROUND Preeclampsia is a serious pregnancy complication affecting 5% to 8% of pregnancies globally. preeclampsia is a leading cause of maternal and neonatal morbidity and death. Despite its prevalence, the underlying mechanisms of preeclampsia remain unclear. This study investigated the role of vasorin in preeclampsia pathogenesis by examining its levels in extracellular vesicles (EVs) and effects on vascular function. METHODS AND RESULTS We conducted unbiased proteomics on urine-derived EVs from women with severe preeclampsia and normotensive pregnancies, identifying differentially abundant proteins. Vasorin expression levels were measured in urinary EVs, plasma EVs, and placental tissue. EVs were generated from human and murine placental explants. Vascular functions were assessed using murine aortic rings and human aortic endothelial cells. Vasorin expression was manipulated in human aortic endothelial cells via overexpression and knockdown followed by RNA sequencing. One hundred twenty proteins showed ≥±1.5-fold regulation (P<0.05) between severe preeclampsia and NTP. Vasorin levels decreased in severe preeclampsia in urinary EVs, plasma EVs, and placental tissue. Vasorin levels increased with gestational age in murine pregnancy and were diminished in a murine model of preeclampsia. Severe preeclampsia and murine preeclampsia EVs impaired human aortic endothelial cell migration and inhibited murine aortic ring vasorelaxation. Vasorin overexpression counteracted these effects. RNA sequencing showed that vasorin manipulation in human aortic endothelial cells differentially regulated hundreds of genes linked to vasculogenesis, proliferation, migration, and apoptosis. CONCLUSIONS The data suggest that vasorin, delivered to the endothelium via EVs, regulates vascular function and that the loss of EV vasorin may be one of the mechanistic drivers of preeclampsia.
Collapse
Affiliation(s)
- Saravanakumar Murugesan
- Department of Anesthesiology and Perioperative Medicine, Division of Molecular and Translational Biomedicine, School of Medicine University of Alabama at Birmingham Birmingham AL USA
| | - Dylan R Addis
- Department of Anesthesiology and Perioperative Medicine, Division of Molecular and Translational Biomedicine, School of Medicine University of Alabama at Birmingham Birmingham AL USA
| | - Hanna Hussey
- Department of Anesthesiology and Perioperative Medicine, Division of Molecular and Translational Biomedicine, School of Medicine University of Alabama at Birmingham Birmingham AL USA
| | - Mark F Powell
- Department of Anesthesiology and Perioperative Medicine, Division of Molecular and Translational Biomedicine, School of Medicine University of Alabama at Birmingham Birmingham AL USA
| | - Lakshmi Saravanakumar
- Department of Anesthesiology and Perioperative Medicine, Division of Molecular and Translational Biomedicine, School of Medicine University of Alabama at Birmingham Birmingham AL USA
| | - Adam B Sturdivant
- Department of Anesthesiology and Perioperative Medicine, Division of Molecular and Translational Biomedicine, School of Medicine University of Alabama at Birmingham Birmingham AL USA
| | - Rachel G Sinkey
- Department of Obstetrics and Gynecology, Division of Maternal-Fetal Medicine, School of Medicine University of Alabama at Birmingham Birmingham AL USA
| | - Michelle D Tubinis
- Department of Anesthesiology and Perioperative Medicine, Division of Molecular and Translational Biomedicine, School of Medicine University of Alabama at Birmingham Birmingham AL USA
| | - Zachary R Massey
- Department of Anesthesiology and Perioperative Medicine, Division of Molecular and Translational Biomedicine, School of Medicine University of Alabama at Birmingham Birmingham AL USA
| | - Chelsi Patton
- Department of Anesthesiology and Perioperative Medicine, Division of Molecular and Translational Biomedicine, School of Medicine University of Alabama at Birmingham Birmingham AL USA
| | - James A Mobley
- Department of Anesthesiology and Perioperative Medicine, Division of Molecular and Translational Biomedicine, School of Medicine University of Alabama at Birmingham Birmingham AL USA
| | - Alan N Tita
- Department of Obstetrics and Gynecology, Division of Maternal-Fetal Medicine, School of Medicine University of Alabama at Birmingham Birmingham AL USA
| | - Tamas Jilling
- Department of Pediatrics, Division of Neonatology University of Alabama at Birmingham Birmingham AL USA
| | - Dan E Berkowitz
- Department of Anesthesiology and Perioperative Medicine, Division of Molecular and Translational Biomedicine, School of Medicine University of Alabama at Birmingham Birmingham AL USA
| |
Collapse
|
4
|
Álvarez-Aznar A, Desai M, Orlich MM, Vázquez-Liébanas E, Adams RH, Brakebusch C, Gaengel K. Cdc42 is crucial for mural cell migration, proliferation and patterning of the retinal vasculature. Vascul Pharmacol 2025; 159:107472. [PMID: 39971261 DOI: 10.1016/j.vph.2025.107472] [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: 09/30/2024] [Revised: 02/12/2025] [Accepted: 02/16/2025] [Indexed: 02/21/2025]
Abstract
AIMS Mural cells constitute the outer lining of blood vessels and are essential for vascular development and function. Mural cell loss or malfunction has been associated with numerous diseases including diabetic retinopathy, stroke and amyotrophic lateral sclerosis. In this work, we investigate the role of CDC42 in mural cells in vivo, using the developing mouse retina as a model. METHODS In this study, we generated a mouse model for Cdc42 deletion in mural cells by crossing Pdgfrb-CreERT2 mice with Cdc42flox/flox mice. This model (Cdc42iΔMC) allowed us to investigate the role of CDC42 in pericytes and smooth muscle cells in the developing and adult retinal vasculature. RESULTS We find that, during postnatal development, CDC42 is required in both, pericytes and smooth muscle cells to maintain proper cell morphology, mural cell coverage and distribution. During retinal angiogenesis, Cdc42-depleted pericytes lag behind the sprouting front and exhibit decreased proliferation. Consequently, capillaries at the sprouting front remain pericyte deprived, become dilated and are prone to increased vascular leakage. In addition, arteries and arterioles deviate from their normal growth directions and trajectory. While in the adult retina, mural cell coverage normalizes and pericytes adopt a normal morphology, smooth muscle cell morphologies remain abnormal and arteriolar branching angles are markedly reduced. CONCLUSIONS Our findings demonstrate that CDC42 is required for mural cell migration and proliferation and suggest that mural cells are essential for normal morphogenesis and patterning of the developing retinal vasculature.
Collapse
Affiliation(s)
- Alberto Álvarez-Aznar
- Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala University, Uppsala, Sweden
| | - Malavika Desai
- Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala University, Uppsala, Sweden
| | - Michael M Orlich
- Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala University, Uppsala, Sweden; Institute of Clinical Chemistry and Laboratory Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Elisa Vázquez-Liébanas
- Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala University, Uppsala, Sweden
| | - Ralf H Adams
- Max Plank Institute for Molecular Medicine, Department of Tissue Morphogenesis, Münster, Germany
| | | | - Konstantin Gaengel
- Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala University, Uppsala, Sweden.
| |
Collapse
|
5
|
Otsuka K, Morita A, Kashihara T, Nakahara T. Pharmacological depletion of pericytes induces diabetic retinopathy-like abnormal blood vessels in neonatal rat retina. Exp Eye Res 2025; 251:110243. [PMID: 39826860 DOI: 10.1016/j.exer.2025.110243] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Revised: 12/26/2024] [Accepted: 01/15/2025] [Indexed: 01/22/2025]
Abstract
Diabetic retinopathy is a major ocular complication associated with diabetes mellitus. Pericyte loss is a hallmark of diabetic retinopathy. The platelet-derived growth factor (PDGF)-B-PDGF receptor-β (PDGFRβ) signaling pathway plays an important role in the proliferation and migration of pericytes. Imatinib, an antineoplastic drug primarily used to treat chronic myelogenous leukemia, inhibits the PDGFRβ tyrosine kinase. In this study, we aimed to determine the time-course of pathological changes in the retinal vasculature following pharmacological depletion of pericytes with imatinib. Rats were injected with imatinib once daily for 1, 2, or 4 days starting on postnatal day (P) 4. The distribution of endothelial cells and pericytes in the retina was assessed at P4, P5, P6, P8, and P11. Single and multiple injections of imatinib (100 mg/kg) significantly decreased the pericyte coverage within the retinal capillaries on the day after the completion of each injection protocol. After pericyte coverage decreased, endothelial cell degeneration and microaneurysm formation were initiated. Following the elimination of the inhibitory effect of imatinib on the PDGFRβ signaling pathway, the pericyte coverage returned to control levels but structural abnormalities of the retinal vasculature with microaneurysms and dense capillaries were observed. Vascular pathological features are similar to those of the early clinical manifestations of diabetic retinopathy. Therefore, these rats could serve as animal models to study the mechanisms underlying the pathological changes that occur after pericyte loss in diabetic retinopathy.
Collapse
Affiliation(s)
- Kenta Otsuka
- Department of Molecular Pharmacology, Kitasato University School of Pharmaceutical Sciences, 5-9-1 Shirokane Minato-ku, Tokyo, 108-8641, Japan
| | - Akane Morita
- Department of Molecular Pharmacology, Kitasato University School of Pharmaceutical Sciences, 5-9-1 Shirokane Minato-ku, Tokyo, 108-8641, Japan
| | - Toshihide Kashihara
- Department of Molecular Pharmacology, Kitasato University School of Pharmaceutical Sciences, 5-9-1 Shirokane Minato-ku, Tokyo, 108-8641, Japan
| | - Tsutomu Nakahara
- Department of Molecular Pharmacology, Kitasato University School of Pharmaceutical Sciences, 5-9-1 Shirokane Minato-ku, Tokyo, 108-8641, Japan.
| |
Collapse
|
6
|
Yu M, Jiang WJ, Yu M, Zhou Z, Wang M, Li L. The Upregulation of IL-1β Induced by Cisplatin Triggers PI3K/AKT/MMP9 Pathway in Pericytes Mediating the Leakage of the Blood Labyrinth Barrier. J Inflamm Res 2025; 18:1191-1205. [PMID: 39911951 PMCID: PMC11794042 DOI: 10.2147/jir.s492292] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2024] [Accepted: 01/09/2025] [Indexed: 02/07/2025] Open
Abstract
Background Blood-labyrinth barrier (BLB) damage has been recognized as a key mechanism underlying cisplatin (CDDP)-induced hearing loss. Inflammation within the cochlea, triggered by CDDP, is a key pathological response. However, the relationship between CDDP-induced inflammation and BLB dysfunction remains elusive. Materials and Methods In vivo and in vitro BLB models were used to explore the inflammatory mechanisms underlying CDDP ototoxicity. C57BL/6J mice were treated with CDDP and IL-1β levels, BLB permeability, and hearing thresholds were assessed using ELISA, histological staining, ABR test and BLB leakage tests. In vitro BLB models, the effect of IL-1β on MMP9 expression, PI3K-AKT pathway activation, and endothelial barrier permeability were examined via Western blot, TEER value test, and FITC extraction analysis. In addition, inhibitors of IL-1β, MMP9, and PI3K-AKT were used to analyze the specific mechanisms. Results After CDDP treatment, IL-1β upregulation in the stria vascularis disrupted tight junctions, increased BLB permeability, and led to hearing loss. Notably, IL-1β inhibition with AS101 attenuated hearing threshold elevation and BLB damage in CDDP-treated mice. Mechanistically, CDDP triggered IL-1β release from endothelial cells. IL-1β promoted MMP9 secretion from pericytes via the PI3K/AKT pathway, leading to disruption of tight junctions. Both MMP9 and PI3K-AKT inhibitors abrogated IL-1β-induced changes. Conclusion Our findings suggest that CDDP initiates a cascade of events starting with IL-1β release from endothelial cells. This release triggers the activation of PI3K/AKT pathway and upregulation of MMP9 expression in pericytes, which increases BLB permeability and leds to hearing loss. IL-1β and the PI3K-AKT pathway are promising therapeutic targets,offering hope for patients with CDDP-induced hearing loss.
Collapse
Affiliation(s)
- Miao Yu
- Department of Physiology, Medical College of Jiaxing University, Jiaxing, Zhejiang, 314000, People’s Republic of China
- Department of Physiology, Medical College of Shihezi University, Shihezi, Xinjiang, 832000, People’s Republic of China
| | - Wen-Jun Jiang
- The Second People’s Hospital of Li Shui, Li Shui, Zhejiang, 323000, People’s Republic of China
- Department of Physiology, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, 310051, People’s Republic of China
| | - Meng Yu
- Department of Physiology, Medical College of Jiaxing University, Jiaxing, Zhejiang, 314000, People’s Republic of China
| | - Zan Zhou
- Department of Physiology, Medical College of Jiaxing University, Jiaxing, Zhejiang, 314000, People’s Republic of China
- Department of Physiology, Medical College of Shihezi University, Shihezi, Xinjiang, 832000, People’s Republic of China
| | - Min Wang
- Department of Physiology, Medical College of Jiaxing University, Jiaxing, Zhejiang, 314000, People’s Republic of China
| | - Li Li
- Department of Physiology, Medical College of Jiaxing University, Jiaxing, Zhejiang, 314000, People’s Republic of China
| |
Collapse
|
7
|
Huang C, Zhang X, Wu M, Yang C, Ge X, Chen W, Li X, Liu S, Yang S. IL-1β-induced pericyte dysfunction with a secretory phenotype exacerbates retinal microenvironment inflammation via Hes1/STAT3 signaling pathway. Int Immunopharmacol 2025; 144:113611. [PMID: 39612772 DOI: 10.1016/j.intimp.2024.113611] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2024] [Revised: 11/06/2024] [Accepted: 11/07/2024] [Indexed: 12/01/2024]
Abstract
Retinal pericytes are mural cells surrounding capillaries to maintain the integrity of blood-retina barrier and regulate vascular behaviors. Pericyte loss has been considered as the hallmark of diabetic retinopathy (DR), which is a major complication of diabetes and the leading cause of blindness in adults. However, the precise function of pericytes in regulating the retinal microenvironment and the underlying mechanism remains largely unknown. In this study, we observed a secretory phenotype of pericytes with elevated inflammatory cytokines in response to Interleukin-1β (IL-1β), a canonical inflammatory cytokine which significantly increases during the initial stages of diabetic retinopathy. This phenotype is also accompanied by reduced expression of adherent junction proteins and contractile proteins. Paracrine cytokines derived from pericytes further induce the chemotaxis of microglia cells and trigger detrimental changes in endothelial cells, including reduced expression of tight junction protein Occludin and increased apoptosis. Mechanically, the secretion potential in pericytes is partially mediated by Hes1/STAT3 signaling pathway. Moreover, co-injection of stattic, an inhibitor targeting STAT3 activation, could effectively attenuate IL-1β-induced retinal inflammation and microglial activation in retina tissues. Collectively, these findings demonstrate the potential of retinal pericytes as an initial inflammatory sensor prior to their anatomical pathological loss, via undergoing phenotypic changes and secreting paracrine factors to amplify local inflammation and damage endothelial cells in vitro. Furthermore, inhibition of STAT3 activation by inhibitors significantly ameliorates IL-1β-induced retinal inflammation, suggesting STAT3 in retinal pericytes as a promising target for alleviating DR and other IL-1β-induced ocular diseases.
Collapse
Affiliation(s)
- Caoxin Huang
- Xiamen Diabetes Institute, Fujian Province Key Laboratory of Translational Research for Diabetes, Department of Endocrinology and Diabetes, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, China.
| | - Xiaofang Zhang
- Xiamen Diabetes Institute, Fujian Province Key Laboratory of Translational Research for Diabetes, Department of Endocrinology and Diabetes, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, China
| | - Menghua Wu
- Xiamen Diabetes Institute, Fujian Province Key Laboratory of Translational Research for Diabetes, Department of Endocrinology and Diabetes, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, China
| | - Chen Yang
- Xiamen Diabetes Institute, Fujian Province Key Laboratory of Translational Research for Diabetes, Department of Endocrinology and Diabetes, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, China
| | - Xilin Ge
- Xiamen Diabetes Institute, Fujian Province Key Laboratory of Translational Research for Diabetes, Department of Endocrinology and Diabetes, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, China
| | - Wenting Chen
- Xiamen Diabetes Institute, Fujian Province Key Laboratory of Translational Research for Diabetes, Department of Endocrinology and Diabetes, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, China
| | - Xuejun Li
- Xiamen Diabetes Institute, Fujian Province Key Laboratory of Translational Research for Diabetes, Department of Endocrinology and Diabetes, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, China
| | - Suhuan Liu
- Xiamen Diabetes Institute, Fujian Province Key Laboratory of Translational Research for Diabetes, Department of Endocrinology and Diabetes, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, China; Research Center for Translational Medicine, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, China.
| | - Shuyu Yang
- Xiamen Diabetes Institute, Fujian Province Key Laboratory of Translational Research for Diabetes, Department of Endocrinology and Diabetes, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, China; Research Studio of Traditional Chinese Medicine, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, Fujian, China.
| |
Collapse
|
8
|
Jeon HH, Huang X, Rojas Cortez L, Sripinun P, Lee JM, Hong JJ, Graves DT. Inflammation and mechanical force-induced bone remodeling. Periodontol 2000 2024. [PMID: 39740162 DOI: 10.1111/prd.12619] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Revised: 09/25/2024] [Accepted: 10/27/2024] [Indexed: 01/02/2025]
Abstract
Periodontitis arises from imbalanced host-microbe interactions, leading to dysbiosis and destructive inflammation. The host's innate and adaptive immune responses produce pro-inflammatory mediators that stimulate destructive events, which cause loss of alveolar bone and connective tissue attachment. There is no consensus on the factors that lead to a conversion from gingivitis to periodontitis, but one possibility is the proximity of the inflammation to the bone, which promotes bone resorption and inhibits subsequent bone formation during coupled bone formation. Conversely, orthodontic tooth movement is triggered by the mechanical force applied to the tooth, resulting in bone resorption on the compression side and new bone formation on the tension side. However, the environment around orthodontic brackets readily retains dental plaque and may contribute to inflammation and bone remodeling. The immune, epithelial, stromal, endothelial and bone cells of the host play an important role in setting the stage for bone remodeling that occurs in both periodontitis and orthodontic tooth movement. Recent advancements in single-cell RNA sequencing have provided new insights into the roles and interactions of different cell types in response to challenges. In this review, we meticulously examine the functions of key cell types such as keratinocytes, leukocytes, stromal cells, osteocytes, osteoblasts, and osteoclasts involved in inflammation- and mechanical force-driven bone remodeling. Moreover, we explore the combined effects of these two conditions: mechanical force-induced bone remodeling combined with periodontal disease (chronic inflammation) and periodontally accelerated osteogenic orthodontics (acute transient inflammation). This comprehensive review enhances our understanding of inflammation- and mechanical force-induced bone remodeling.
Collapse
Affiliation(s)
- Hyeran Helen Jeon
- Department of Orthodontics, School of Dental Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Xin Huang
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, Guangdong, China
- Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou, Guangdong, China
- Department of Periodontics, School of Dental Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Leticia Rojas Cortez
- Department of Periodontics, School of Dental Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Puttipong Sripinun
- Department of Orthodontics, School of Dental Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Department of Orthodontics and Pediatric Dentistry, Faculty of Dentistry, Chiang Mai University, Muang, Chiang Mai, Thailand
| | - Jung-Me Lee
- Division of Nutritional Sciences, College of Human Ecology, Cornell University, Ithaca, New York, USA
| | - Julie J Hong
- Department of Orthodontics, School of Dental Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Department of Periodontics, School of Dental Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Dana T Graves
- Department of Periodontics, School of Dental Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| |
Collapse
|
9
|
Wang Y, Gao S, Cao F, Yang H, Lei F, Hou S. Ocular immune-related diseases: molecular mechanisms and therapy. MedComm (Beijing) 2024; 5:e70021. [PMID: 39611043 PMCID: PMC11604294 DOI: 10.1002/mco2.70021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Revised: 10/05/2024] [Accepted: 10/31/2024] [Indexed: 11/30/2024] Open
Abstract
Ocular immune-related diseases, represent a spectrum of conditions driven by immune system dysregulation, include but not limit to uveitis, diabetic retinopathy, age-related macular degeneration, Graves' ophthalmopathy, etc. The molecular and cellular mechanisms underlying these diseases are typically dysfunctioned immune responses targeting ocular tissues, resulting in inflammation and tissue damage. Recent advances have further elucidated the pivotal role of different immune responses in the development, progression, as well as management of various ocular immune diseases. However, there is currently a relative lack of connection between the cellular mechanisms and treatments of several immune-related ocular diseases. In this review, we discuss recent findings related to the immunopathogenesis of above-mentioned diseases. In particular, we summarize the different types of immune cells, inflammatory mediators, and associated signaling pathways that are involved in the pathophysiology of above-mentioned ophthalmopathies. Furthermore, we also discuss the future directions of utilizing anti-inflammatory regime in the management of these diseases. This will facilitate a better understanding of the pathogenesis of immune-related ocular diseases and provide new insights for future treatment approaches.
Collapse
Affiliation(s)
- Yakun Wang
- The First Affiliated Hospital of Chongqing Medical UniversityChongqingChina
- Beijing Institute of OphthalmologyBeijing Tongren Eye CenterBeijing Tongren Hospital, Beijing Ophthalmology & Visual Sciences Key Laboratory, Capital Medical UniversityBeijingChina
| | - Shangze Gao
- Beijing Institute of OphthalmologyBeijing Tongren Eye CenterBeijing Tongren Hospital, Beijing Ophthalmology & Visual Sciences Key Laboratory, Capital Medical UniversityBeijingChina
| | - Fan Cao
- Beijing Institute of OphthalmologyBeijing Tongren Eye CenterBeijing Tongren Hospital, Beijing Ophthalmology & Visual Sciences Key Laboratory, Capital Medical UniversityBeijingChina
| | - Hui Yang
- Beijing Institute of OphthalmologyBeijing Tongren Eye CenterBeijing Tongren Hospital, Beijing Ophthalmology & Visual Sciences Key Laboratory, Capital Medical UniversityBeijingChina
| | - Fengyang Lei
- Beijing Institute of OphthalmologyBeijing Tongren Eye CenterBeijing Tongren Hospital, Beijing Ophthalmology & Visual Sciences Key Laboratory, Capital Medical UniversityBeijingChina
| | - Shengping Hou
- Beijing Institute of OphthalmologyBeijing Tongren Eye CenterBeijing Tongren Hospital, Beijing Ophthalmology & Visual Sciences Key Laboratory, Capital Medical UniversityBeijingChina
| |
Collapse
|
10
|
Wang S, Tong S, Jin X, Li N, Dang P, Sui Y, Liu Y, Wang D. Single-cell RNA sequencing analysis of the retina under acute high intraocular pressure. Neural Regen Res 2024; 19:2522-2531. [PMID: 38526288 PMCID: PMC11090430 DOI: 10.4103/1673-5374.389363] [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: 05/24/2023] [Revised: 07/27/2023] [Accepted: 09/13/2023] [Indexed: 03/26/2024] Open
Abstract
JOURNAL/nrgr/04.03/01300535-202419110-00032/figure1/v/2024-03-08T184507Z/r/image-tiff High intraocular pressure causes retinal ganglion cell injury in primary and secondary glaucoma diseases, yet the molecular landscape characteristics of retinal cells under high intraocular pressure remain unknown. Rat models of acute hypertension ocular pressure were established by injection of cross-linked hyaluronic acid hydrogel (Healaflow®). Single-cell RNA sequencing was then used to describe the cellular composition and molecular profile of the retina following high intraocular pressure. Our results identified a total of 12 cell types, namely retinal pigment epithelial cells, rod-photoreceptor cells, bipolar cells, Müller cells, microglia, cone-photoreceptor cells, retinal ganglion cells, endothelial cells, retinal progenitor cells, oligodendrocytes, pericytes, and fibroblasts. The single-cell RNA sequencing analysis of the retina under acute high intraocular pressure revealed obvious changes in the proportions of various retinal cells, with ganglion cells decreased by 23%. Hematoxylin and eosin staining and TUNEL staining confirmed the damage to retinal ganglion cells under high intraocular pressure. We extracted data from retinal ganglion cells and analyzed the retinal ganglion cell cluster with the most distinct expression. We found upregulation of the B3gat2 gene, which is associated with neuronal migration and adhesion, and downregulation of the Tsc22d gene, which participates in inhibition of inflammation. This study is the first to reveal molecular changes and intercellular interactions in the retina under high intraocular pressure. These data contribute to understanding of the molecular mechanism of retinal injury induced by high intraocular pressure and will benefit the development of novel therapies.
Collapse
Affiliation(s)
- Shaojun Wang
- Division of Ophthalmology, The Third Medical Center of PLA General Hospital, Beijing, China
| | - Siti Tong
- Division of Ophthalmology, The Third Medical Center of PLA General Hospital, Beijing, China
| | - Xin Jin
- Division of Ophthalmology, The Third Medical Center of PLA General Hospital, Beijing, China
| | - Na Li
- Division of Ophthalmology, The Third Medical Center of PLA General Hospital, Beijing, China
| | - Pingxiu Dang
- Division of Ophthalmology, The Third Medical Center of PLA General Hospital, Beijing, China
| | - Yang Sui
- Division of Ophthalmology, The Third Medical Center of PLA General Hospital, Beijing, China
| | - Ying Liu
- Department of Ophthalmology, Beijing Rehabilitation Hospital, Capital Medical University, Beijing, China
| | - Dajiang Wang
- Division of Ophthalmology, The Third Medical Center of PLA General Hospital, Beijing, China
| |
Collapse
|
11
|
Wang S, Bao N, Li M, Liu D, Tao L. Ets2 Exacerbates Diabetic Retinopathy by Aggravating the Proliferation of Endothelial Cells and Inflammatory Response. Biochem Genet 2024:10.1007/s10528-024-10938-8. [PMID: 39432129 DOI: 10.1007/s10528-024-10938-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Accepted: 10/05/2024] [Indexed: 10/22/2024]
Abstract
Proliferative diabetic retinopathy (PDR), the most common type of diabetic retinopathy, is a main cause of visual and impairment blindness. Abnormal neovascularization, endothelial dysfunction, and vascular inflammation are important mechanisms for the development of PDR. Ets2 regulates angiogenesis-related genes and inflammation, however, the effect of Ets2 in PDR procession has not been clarified. Thus, this study is performed to investigate whether Ets2 exerts key functions in PDR. In this study, 10-week-old mice were used for establishing STZ-induced diabetic mice, and Ets2 expression was analyzed in retina tissues. Besides, newborn mice were applied to construct oxygen-induced retinopathy (OIR) models. The Ets2 expression, oxidative stress, and inflammation were detected in retina tissues. We found that Ets2 was highly expressed in retina tissues both in diabetic mice and OIR mice. Oxidative stress and inflammatory processes are two factors contributing to the pathogenesis of PDR. In retinal tissues of OIR mice, Ets2 knockdown inhibited expression of inflammatory mediators VEGFA, IL-6, and IL-8, and biomarkers of oxidative stress MCP-1, VCAM-1, and iNOS. ROS production was also inhibited by silencing Ets2. Ets2 deficiency inhibited endothelial cell proliferation in the retina. Furthermore, Ets2 knockdown contributed to suppressing the expression of angiogenesis-related genes VEGFA, JUNB, MMP-9, Tie2, Ang-2, and EphB4. Our study highlights that Ets2 accelerates PDR procession by promoting the proliferation of endothelial cells, oxidative stress, and inflammation, which provides a novel target against PDR.
Collapse
Affiliation(s)
- Song Wang
- Department of Ophthalmology, the Second Affiliated Hospital of Anhui Medical University, No. 678 Furong Road, Hefei, 230601, Anhui, China
| | - Ning Bao
- Department of Ophthalmology, the Second Affiliated Hospital of Anhui Medical University, No. 678 Furong Road, Hefei, 230601, Anhui, China
| | - Mohan Li
- Department of Ophthalmology, the Second Affiliated Hospital of Anhui Medical University, No. 678 Furong Road, Hefei, 230601, Anhui, China
| | - Dongwei Liu
- Department of Ophthalmology, the Second Affiliated Hospital of Anhui Medical University, No. 678 Furong Road, Hefei, 230601, Anhui, China
| | - Liming Tao
- Department of Ophthalmology, the Second Affiliated Hospital of Anhui Medical University, No. 678 Furong Road, Hefei, 230601, Anhui, China.
| |
Collapse
|
12
|
Xu W, Cui LJ, Yang XY, Cui XY, Guo J, Xu GX. Impaired pericyte-Müller glia interaction via PDGFRβ suppression aggravates photoreceptor loss in a rodent model of light-induced retinal injury. Int J Ophthalmol 2024; 17:1800-1808. [PMID: 39430007 PMCID: PMC11422380 DOI: 10.18240/ijo.2024.10.05] [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: 05/28/2023] [Accepted: 06/21/2024] [Indexed: 10/22/2024] Open
Abstract
AIM To investigate the involvement of pericyte-Müller glia interaction in retinal damage repair and assess the influence of suppressing the platelet-derived growth factor receptor β (PDGFRβ) signaling pathway in retinal pericytes on photoreceptor loss and Müller glial response. METHODS Sprague-Dawley rats were exposed to intense light to induce retinal injury. Neutralizing antibody against PDGFRβ were deployed to block the signaling pathway in retinal pericytes through intravitreal injection. Retinal histology and Müller glial reaction were assessed following light injury. In vitro, normal and PDGFRβ-blocked retinal pericytes were cocultured with Müller cell line (rMC-1) to examine morphological and protein expression changes upon supplementation with light-injured supernatants of homogenized retinas (SHRs). RESULTS PDGFRβ blockage 24h prior to intense light exposure resulted in a significant exacerbation of photoreceptor loss. The upregulation of GFAP and p-STAT3, observed after intense light exposure, was significantly inhibited in the PDGFRβ blockage group. Further upregulation of cytokines monocyte chemoattractant protein 1 (MCP-1) and interleukin-1β (IL-1β) was also observed following PDGFRβ inhibition. In the in vitro coculture system, the addition of light-injured SHRs induced pericyte deformation and upregulation of proliferating cell nuclear antigen (PCNA) expression, while Müller cells exhibited neuron-like morphology and expressed Nestin. However, PDGFRβ blockage in retinal pericytes abolished these cellular responses to light-induced damage, consistent with the in vivo PDGFRβ blockage findings. CONCLUSION Pericyte-Müller glia interaction plays a potential role in the endogenous repair process of retinal injury. Impairment of this interaction exacerbates photoreceptor degeneration in light-induced retinal injury.
Collapse
Affiliation(s)
- Wei Xu
- Department of Ophthalmology, the First Affiliated Hospital of Fujian Medical University, Fuzhou 350001, Fujian Province, China
- Department of Ophthalmology, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital of Fujian Medical University, Fuzhou 350212, Fujian Province, China
- Fujian Institute of Ophthalmology, Fuzhou 350001, Fujian Province, China
| | - Li-Jin Cui
- Department of Ophthalmology, the First Affiliated Hospital of Fujian Medical University, Fuzhou 350001, Fujian Province, China
- Department of Ophthalmology, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital of Fujian Medical University, Fuzhou 350212, Fujian Province, China
- Fujian Institute of Ophthalmology, Fuzhou 350001, Fujian Province, China
| | - Xiao-Ying Yang
- Department of Ophthalmology, the First Affiliated Hospital of Fujian Medical University, Fuzhou 350001, Fujian Province, China
| | - Xiao-Yuan Cui
- Department of Ophthalmology, the First Affiliated Hospital of Fujian Medical University, Fuzhou 350001, Fujian Province, China
| | - Jian Guo
- Department of Ophthalmology, the First Affiliated Hospital of Fujian Medical University, Fuzhou 350001, Fujian Province, China
- Department of Ophthalmology, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital of Fujian Medical University, Fuzhou 350212, Fujian Province, China
- Fujian Institute of Ophthalmology, Fuzhou 350001, Fujian Province, China
| | - Guo-Xing Xu
- Department of Ophthalmology, the First Affiliated Hospital of Fujian Medical University, Fuzhou 350001, Fujian Province, China
- Department of Ophthalmology, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital of Fujian Medical University, Fuzhou 350212, Fujian Province, China
- Fujian Institute of Ophthalmology, Fuzhou 350001, Fujian Province, China
| |
Collapse
|
13
|
Mosavi-Hecht RM, Yang P, Heyer B, Rosenberg CR, White E, Berry EG, Duvoisin RM, Morgans CW. Case report: Longitudinal evaluation and treatment of a melanoma-associated retinopathy patient. Front Med (Lausanne) 2024; 11:1445180. [PMID: 39318594 PMCID: PMC11420136 DOI: 10.3389/fmed.2024.1445180] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Accepted: 08/26/2024] [Indexed: 09/26/2024] Open
Abstract
Melanoma-associated retinopathy (MAR) is a paraneoplastic syndrome associated with cutaneous metastatic melanoma in which patients develop vision deficits that include reduced night vision, poor contrast sensitivity, and photopsia. MAR is caused by autoantibodies targeting TRPM1, an ion channel found in melanocytes and retinal ON-bipolar cells (ON-BCs). The visual symptoms arise when TRPM1 autoantibodies enter ON-BCs and block the function of TRPM1, thus detection of TRPM1 autoantibodies in patient serum is a key criterion in diagnosing MAR. Electroretinograms are used to measure the impact of TRPM1 autoantibodies on ON-BC function and represent another important diagnostic tool for MAR. To date, MAR case reports have included one or both diagnostic components, but only for a single time point in the course of a patient's disease. Here, we report a case of MAR supported by longitudinal analysis of serum autoantibody detection, visual function, ocular inflammation, vascular integrity, and response to slow-release intraocular corticosteroids. Integrating these data with the patient's oncological and ophthalmological records reveals novel insights regarding MAR pathogenesis, progression, and treatment, which may inform new research and expand our collective understanding of the disease. In brief, we find TRPM1 autoantibodies can disrupt vision even when serum levels are barely detectable by western blot and immunohistochemistry; intraocular dexamethasone treatment alleviates MAR visual symptoms despite high levels of circulating TRPM1 autoantibodies, implicating antibody access to the retina as a key factor in MAR pathogenesis. Elevated inflammatory cytokine levels in the patient's eyes may be responsible for the observed damage to the blood-retinal barrier and subsequent entry of autoantibodies into the retina.
Collapse
Affiliation(s)
- Ryan M. Mosavi-Hecht
- Department of Chemical Physiology and Biochemistry, Oregon Health & Science University, Portland, OR, United States
| | - Paul Yang
- Casey Eye Institute, Oregon Health & Science University, Portland, OR, United States
| | - Barrett Heyer
- Department of Chemical Physiology and Biochemistry, Oregon Health & Science University, Portland, OR, United States
| | | | - Elizabeth White
- Casey Eye Institute, Oregon Health & Science University, Portland, OR, United States
| | - Elizabeth G. Berry
- Department of Dermatology, Oregon Health & Science University, Portland, OR, United States
| | - Robert M. Duvoisin
- Department of Chemical Physiology and Biochemistry, Oregon Health & Science University, Portland, OR, United States
- Casey Eye Institute, Oregon Health & Science University, Portland, OR, United States
| | - Catherine W. Morgans
- Department of Chemical Physiology and Biochemistry, Oregon Health & Science University, Portland, OR, United States
- Casey Eye Institute, Oregon Health & Science University, Portland, OR, United States
| |
Collapse
|
14
|
Hu A, Schmidt MHH, Heinig N. Microglia in retinal angiogenesis and diabetic retinopathy. Angiogenesis 2024; 27:311-331. [PMID: 38564108 PMCID: PMC11303477 DOI: 10.1007/s10456-024-09911-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Accepted: 02/18/2024] [Indexed: 04/04/2024]
Abstract
Diabetic retinopathy has a high probability of causing visual impairment or blindness throughout the disease progression and is characterized by the growth of new blood vessels in the retina at an advanced, proliferative stage. Microglia are a resident immune population in the central nervous system, known to play a crucial role in regulating retinal angiogenesis in both physiological and pathological conditions, including diabetic retinopathy. Physiologically, they are located close to blood vessels and are essential for forming new blood vessels (neovascularization). In diabetic retinopathy, microglia become widely activated, showing a distinct polarization phenotype that leads to their accumulation around neovascular tufts. These activated microglia induce pathogenic angiogenesis through the secretion of various angiogenic factors and by regulating the status of endothelial cells. Interestingly, some subtypes of microglia simultaneously promote the regression of neovascularization tufts and normal angiogenesis in neovascularization lesions. Modulating the state of microglial activation to ameliorate neovascularization thus appears as a promising potential therapeutic approach for managing diabetic retinopathy.
Collapse
Affiliation(s)
- Aiyan Hu
- Institute of Anatomy, Medical Faculty Carl Gustav Carus, Technische Universität Dresden School of Medicine, Fetscherstr 74, 01307, Dresden, Germany
| | - Mirko H H Schmidt
- Institute of Anatomy, Medical Faculty Carl Gustav Carus, Technische Universität Dresden School of Medicine, Fetscherstr 74, 01307, Dresden, Germany.
| | - Nora Heinig
- Institute of Anatomy, Medical Faculty Carl Gustav Carus, Technische Universität Dresden School of Medicine, Fetscherstr 74, 01307, Dresden, Germany.
| |
Collapse
|
15
|
Yamato M, Kato N, Yamada KI, Inoguchi T. The Early Pathogenesis of Diabetic Retinopathy and Its Attenuation by Sodium-Glucose Transporter 2 Inhibitors. Diabetes 2024; 73:1153-1166. [PMID: 38608284 PMCID: PMC11208076 DOI: 10.2337/db22-0970] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Accepted: 04/02/2024] [Indexed: 04/14/2024]
Abstract
The early pathogenetic mechanism of diabetic retinopathy (DR) and its treatment remain unclear. Therefore, we used streptozotocin-induced diabetic mice to investigate the early pathogenic alterations in DR and the protective effect of sodium-glucose cotransporter 2 (SGLT2) inhibitors against these alterations. Retinal vascular leakage was assessed by dextran fluorescence angiography. Retinal thickness and vascular leakage were increased 2 and 4 weeks after onset of diabetes, respectively. Immunostaining showed that morphological change of microglia (amoeboid form) was observed at 2 weeks. Subsequently, increased angiopoietin-2 expression, simultaneous loss of pericytes and endothelial cells, decreased vessel density, retinal hypoxia, and increased vascular endothelial growth factor (VEGF)-A/VEGF receptor system occurred at 4 weeks. SGLT2 inhibitors (luseogliflozin and ipragliflozin) had a significant protective effect on retinal vascular leakage and retinal thickness at a low dose that did not show glucose-lowering effects. Furthermore, both inhibitors at this dose attenuated microglia morphological changes and these early pathogenic alterations in DR. In vitro study showed both inhibitors attenuated the lipopolysaccharide-induced activation of primary microglia, along with morphological changes toward an inactive form, suggesting the direct inhibitory effect of SGLT2 inhibitors on microglia. In summary, SGLT2 inhibitors may directly prevent early pathogenic mechanisms, thereby potentially playing a role in preventing DR. ARTICLE HIGHLIGHTS
Collapse
Affiliation(s)
- Mayumi Yamato
- Physical Chemistry for Life Science Laboratory, Faculty of Pharmaceutical Sciences, Kyushu University, Fukuoka, Japan
| | - Nao Kato
- Physical Chemistry for Life Science Laboratory, Faculty of Pharmaceutical Sciences, Kyushu University, Fukuoka, Japan
| | - Ken-ichi Yamada
- Physical Chemistry for Life Science Laboratory, Faculty of Pharmaceutical Sciences, Kyushu University, Fukuoka, Japan
| | - Toyoshi Inoguchi
- Fukuoka City Health Promotion Support Center, Fukuoka City Medical Association, Fukuoka, Japan
| |
Collapse
|
16
|
Sheng X, Zhang C, Zhao J, Xu J, Zhang P, Ding Q, Zhang J. Microvascular destabilization and intricated network of the cytokines in diabetic retinopathy: from the perspective of cellular and molecular components. Cell Biosci 2024; 14:85. [PMID: 38937783 PMCID: PMC11212265 DOI: 10.1186/s13578-024-01269-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Accepted: 06/19/2024] [Indexed: 06/29/2024] Open
Abstract
Microvascular destabilization is the primary cause of the inner blood-retinal barrier (iBRB) breakdown and increased vascular leakage in diabetic retinopathy (DR). Microvascular destabilization results from the combinational effects of increased levels of growth factors and cytokines, involvement of inflammation, and the changed cell-to-cell interactions, especially the loss of endothelial cells and pericytes, due to hyperglycemia and hypoxia. As the manifestation of microvascular destabilization, the fluid transports via paracellular and transcellular routes increase due to the disruption of endothelial intercellular junctional complexes and/or the altered caveolar transcellular transport across the retinal vascular endothelium. With diabetes progression, the functional and the structural changes of the iBRB components, including the cellular and noncellular components, further facilitate and aggravate microvascular destabilization, resulting in macular edema, the neuroretinal damage and the dysfunction of retinal inner neurovascular unit (iNVU). Although there have been considerable recent advances towards a better understanding of the complex cellular and molecular network underlying the microvascular destabilization, some still remain to be fully elucidated. Recent data indicate that targeting the intricate signaling pathways may allow to against the microvascular destabilization. Therefore, efforts have been made to better clarify the cellular and molecular mechanisms that are involved in the microvascular destabilization in DR. In this review, we discuss: (1) the brief introduction of DR and microvascular destabilization; (2) the cellular and molecular components of iBRB and iNVU, and the breakdown of iBRB; (3) the matrix and cell-to-cell contacts to maintain microvascular stabilization, including the endothelial glycocalyx, basement membrane, and various cell-cell interactions; (4) the molecular mechanisms mediated cell-cell contacts and vascular cell death; (5) the altered cytokines and signaling pathways as well as the intricate network of the cytokines involved in microvascular destabilization. This comprehensive review aimed to provide the insights for microvascular destabilization by targeting the key molecules or specific iBRB cells, thus restoring the function and structure of iBRB and iNVU, to treat DR.
Collapse
Affiliation(s)
- Xia Sheng
- People's Hospital of Huangdao District, Qingdao, Shandong Province, China
| | - Chunmei Zhang
- People's Hospital of Huangdao District, Qingdao, Shandong Province, China
| | - Jiwei Zhao
- People's Hospital of Huangdao District, Qingdao, Shandong Province, China
| | - Jianping Xu
- People's Hospital of Huangdao District, Qingdao, Shandong Province, China.
| | - Peng Zhang
- People's Hospital of Huangdao District, Qingdao, Shandong Province, China.
| | - Quanju Ding
- People's Hospital of Huangdao District, Qingdao, Shandong Province, China.
| | - Jingfa Zhang
- Department of Ophthalmology, Shanghai General Hospital (Shanghai First People's Hospital), Shanghai Jiao Tong University School of Medicine, Shanghai, China.
- 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, National Clinical Research Center for Eye Diseases, Shanghai, China.
- The International Eye Research Institute of The Chinese University of Hong Kong (Shenzhen), Shenzhen, China.
- C-MER (Shenzhen) Dennis Lam Eye Hospital, Shenzhen, China.
- C-MER International Eye Care Group, C-MER Dennis Lam & Partners Eye Center, Hong Kong, China.
| |
Collapse
|
17
|
Murugesan S, Addis DR, Hussey H, Powell MF, Saravanakumar L, Sturdivant AB, Sinkey RG, Tubinis MD, Massey ZR, Mobley JA, Tita AN, Jilling T, Berkowitz DE. Decreased Extracellular Vesicle Vasorin in Severe Preeclampsia Plasma Mediates Endothelial Dysfunction. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.24.600441. [PMID: 38979275 PMCID: PMC11230191 DOI: 10.1101/2024.06.24.600441] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/10/2024]
Abstract
Background Preeclampsia (PE) is a serious pregnancy complication affecting 5-8% of pregnancies globally. It is a leading cause of maternal and neonatal morbidity and mortality. Despite its prevalence, the underlying mechanisms of PE remain unclear. This study aimed to determine the potential role of vasorin (VASN) in PE pathogenesis by investigating its levels in extracellular vesicles (EV) and its effects on vascular function. Methods & Results We conducted unbiased proteomics on urine-derived EV from severe PE (sPE) and normotensive pregnant women (NTP), identifying differential protein abundances. Out of one hundred and twenty proteins with ≥ ±1.5-fold regulation at P<0.05 between sPE and NTP, we focused on Vasorin (VASN), which is downregulated in sPE in urinary EV, in plasma EV and in the placenta and is a known regulator of vascular function. We generated EV with high VASN content from both human and murine placenta explants (Plex EV), which recapitulated disease-state-dependent effects on vascular function observed when treating murine aorta rings (MAR) or human aortic endothelial cells (HAEC) with murine or human plasma-derived EV. In normal murine pregnancy, VASN increases with gestational age (GA), and VASN is decreased in plasma EV, in placenta tissue and in Plex EV after intravenous administration of adenovirus encoding short FMS-like tyrosine kinase 1 (sFLT-1), a murine model of PE (murine-PE). VASN is decreased in plasma EV, in placenta tissue and in EV isolated from conditioned media collected from placenta explants (Plex EV) in patients with sPE as compared to NTP. Human sPE and murine-PE plasma EV and Plex EV impair migration, tube formation, and induces apoptosis in human aortic endothelial cells (HAEC) and inhibit acetylcholine-induced vasorelaxation in murine vascular rings (MAR). VASN over-expression counteracts the effects of sPE EV treatment in HAEC and MAR. RNA sequencing revealed that over-expression or knock down of VASN in HAEC results in contrasting effects on transcript levels of hundreds of genes associated with vasculogenesis, endothelial cell proliferation, migration and apoptosis. Conclusions The data suggest that VASN, delivered to the endothelium via EV, regulates vascular function and that the loss of EV VASN may be one of the mechanistic drivers of PE. CLINICAL PERSPECTIVE What is NewVASN in circulating plasma EV in sPE is reduced compared with VASN content in plasma EV of gestational age-matched pregnant women.VASN is encapsulated and transported in EV and plays a pro-angiogenic role during pregnancy.VASN should be explored both for its pro-angiogenic mechanistic role and as a novel biomarker and potential predictive diagnostic marker for the onset and severity of PE.What Are the Clinical Implications?VASN plays a role in maintaining vascular health and the normal adaptive cardiovascular response in pregnancy. A decrease of VASN is observed in sPE patients contributing to cardiovascular maladaptation.Strategies to boost diminished VASN levels and/or to pharmacologically manipulate mechanisms downstream of VASN may be explored for potential therapeutic benefit in PE.The decrease in EV-associated VASN could potentially be used as a (predictive) biomarker for PE.
Collapse
|
18
|
Mosavi-Hecht R, Yang P, Heyer B, Rosenberg CR, White E, Berry EG, Duvoisin RM, Morgans CW. Case Report: Longitudinal Evaluation and Treatment of a Melanoma-Associated Retinopathy Patient. RESEARCH SQUARE 2024:rs.3.rs-4595829. [PMID: 38946992 PMCID: PMC11213222 DOI: 10.21203/rs.3.rs-4595829/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/02/2024]
Abstract
Melanoma-associated retinopathy (MAR) is a paraneoplastic syndrome associated with cutaneous metastatic melanoma in which patients develop vision deficits that include reduced night vision, poor contrast sensitivity, and photopsia. MAR is caused by autoantibodies targeting TRPM1, an ion channel found in melanocytes and retinal ON-bipolar cells (ON-BCs). The visual symptoms arise when TRPM1 autoantibodies enter ON-BCs and block the function of TRPM1, thus detection of TRPM1 autoantibodies in patient serum is a key criterion in diagnosing MAR. Electroretinograms are used to measure the impact of TRPM1 autoantibodies on ON-BC function and represent another important diagnostic tool for MAR. To date, MAR case reports have included one or both diagnostic components, but only for a single time point in the course of a patient's disease. Here, we report a case of MAR supported by longitudinal analysis of serum autoantibody detection, visual function, ocular inflammation, vascular integrity, and response to slow-release intraocular corticosteroids. Integrating these data with the patient's oncological and ophthalmological records reveals novel insights regarding MAR pathogenesis, progression, and treatment, which may inform new research and expand our collective understanding of the disease. In brief, we find TRPM1 autoantibodies can disrupt vision even when serum levels are barely detectable by western blot and immunohistochemistry; intraocular dexamethasone treatment alleviates MAR visual symptoms despite high levels of circulating TRPM1 autoantibodies, implicating antibody access to the retina as a key factor in MAR pathogenesis. Elevated inflammatory cytokine levels in the patient's eyes may be responsible for the observed damage to the blood-retinal barrier and subsequent entry of autoantibodies into the retina.
Collapse
Affiliation(s)
| | - Paul Yang
- Oregon Health and Science University, Casey Eye Institute
| | | | | | | | | | | | | |
Collapse
|
19
|
Schonblum A, Ali Naser D, Ovadia S, Egbaria M, Puyesky S, Epshtein A, Wald T, Mercado-Medrez S, Ashery-Padan R, Landsman L. Beneficial islet inflammation in health depends on pericytic TLR/MyD88 signaling. J Clin Invest 2024; 134:e179335. [PMID: 38885342 PMCID: PMC11245159 DOI: 10.1172/jci179335] [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: 01/11/2024] [Accepted: 05/24/2024] [Indexed: 06/20/2024] Open
Abstract
While inflammation is beneficial for insulin secretion during homeostasis, its transformation adversely affects β cells and contributes to diabetes. However, the regulation of islet inflammation for maintaining glucose homeostasis remains largely unknown. Here, we identified pericytes as pivotal regulators of islet immune and β cell function in health. Islets and pancreatic pericytes express various cytokines in healthy humans and mice. To interfere with the pericytic inflammatory response, we selectively inhibited the TLR/MyD88 pathway in these cells in transgenic mice. The loss of MyD88 impaired pericytic cytokine production. Furthermore, MyD88-deficient mice exhibited skewed islet inflammation with fewer cells, an impaired macrophage phenotype, and reduced IL-1β production. This aberrant pericyte-orchestrated islet inflammation was associated with β cell dedifferentiation and impaired glucose response. Additionally, we found that Cxcl1, a pericytic MyD88-dependent cytokine, promoted immune IL-1β production. Treatment with either Cxcl1 or IL-1β restored the mature β cell phenotype and glucose response in transgenic mice, suggesting a potential mechanism through which pericytes and immune cells regulate glucose homeostasis. Our study revealed pericyte-orchestrated islet inflammation as a crucial element in glucose regulation, implicating this process as a potential therapeutic target for diabetes.
Collapse
Affiliation(s)
- Anat Schonblum
- Department of Cell and Development Biology, Faculty of Medical and Health Sciences and
| | - Dunia Ali Naser
- Department of Cell and Development Biology, Faculty of Medical and Health Sciences and
| | - Shai Ovadia
- Department of Human Molecular Genetics and Biochemistry, Faculty of Medical and Health Sciences and Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel
| | - Mohammed Egbaria
- Department of Cell and Development Biology, Faculty of Medical and Health Sciences and
| | - Shani Puyesky
- Department of Cell and Development Biology, Faculty of Medical and Health Sciences and
| | - Alona Epshtein
- Department of Cell and Development Biology, Faculty of Medical and Health Sciences and
| | - Tomer Wald
- Department of Cell and Development Biology, Faculty of Medical and Health Sciences and
| | - Sophia Mercado-Medrez
- Department of Cell and Development Biology, Faculty of Medical and Health Sciences and
| | - Ruth Ashery-Padan
- Department of Human Molecular Genetics and Biochemistry, Faculty of Medical and Health Sciences and Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel
| | - Limor Landsman
- Department of Cell and Development Biology, Faculty of Medical and Health Sciences and
| |
Collapse
|
20
|
Reddy SK, Devi V, Seetharaman ATM, Shailaja S, Bhat KMR, Gangaraju R, Upadhya D. Cell and molecular targeted therapies for diabetic retinopathy. Front Endocrinol (Lausanne) 2024; 15:1416668. [PMID: 38948520 PMCID: PMC11211264 DOI: 10.3389/fendo.2024.1416668] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Accepted: 05/27/2024] [Indexed: 07/02/2024] Open
Abstract
Diabetic retinopathy (DR) stands as a prevalent complication in the eye resulting from diabetes mellitus, predominantly associated with high blood sugar levels and hypertension as individuals age. DR is a severe microvascular complication of both type I and type II diabetes mellitus and the leading cause of vision impairment. The critical approach to combatting and halting the advancement of DR lies in effectively managing blood glucose and blood pressure levels in diabetic patients; however, this is seldom achieved. Both human and animal studies have revealed the intricate nature of this condition involving various cell types and molecules. Aside from photocoagulation, the sole therapy targeting VEGF molecules in the retina to prevent abnormal blood vessel growth is intravitreal anti-VEGF therapy. However, a substantial portion of cases, approximately 30-40%, do not respond to this treatment. This review explores distinctive pathophysiological phenomena of DR and identifiable cell types and molecules that could be targeted to mitigate the chronic changes occurring in the retina due to diabetes mellitus. Addressing the significant research gap in this domain is imperative to broaden the treatment options available for managing DR effectively.
Collapse
Affiliation(s)
- Shivakumar K. Reddy
- Centre for Molecular Neurosciences, Kasturba Medical College, Manipal, Manipal Academy of Higher Education, Manipal, India
| | - Vasudha Devi
- Department of Pharmacology, Kasturba Medical College, Manipal, Manipal Academy of Higher Education, Manipal, India
| | - Amritha T. M. Seetharaman
- Department of Ophthalmology, The University of Tennessee Health Science Center, Memphis, TN, United States
| | - S. Shailaja
- Department of Ophthalmology, Kasturba Medical College, Manipal, Manipal Academy of Higher Education, Manipal, India
| | - Kumar M. R. Bhat
- Department of Anatomy, Kasturba Medical College, Manipal, Manipal Academy of Higher Education, Manipal, India
| | - Rajashekhar Gangaraju
- Department of Ophthalmology, The University of Tennessee Health Science Center, Memphis, TN, United States
- Department of Anatomy & Neurobiology, The University of Tennessee Health Science Center, Memphis, TN, United States
| | - Dinesh Upadhya
- Centre for Molecular Neurosciences, Kasturba Medical College, Manipal, Manipal Academy of Higher Education, Manipal, India
| |
Collapse
|
21
|
Zhang Q, Yan X, Han H, Wang Y, Sun J. Pericyte in retinal vascular diseases: A multifunctional regulator and potential therapeutic target. FASEB J 2024; 38:e23679. [PMID: 38780117 DOI: 10.1096/fj.202302624r] [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: 12/19/2023] [Revised: 04/17/2024] [Accepted: 05/07/2024] [Indexed: 05/25/2024]
Abstract
Retinal vascular diseases (RVDs), in particular diabetic retinopathy, retinal vein occlusion, and retinopathy of prematurity, are leading contributors to blindness. The pathogenesis of RVD involves vessel dilatation, leakage, and occlusion; however, the specific underlying mechanisms remain unclear. Recent findings have indicated that pericytes (PCs), as critical members of the vascular mural cells, significantly contribute to the progression of RVDs, including detachment from microvessels, alteration of contractile and secretory properties, and excessive production of the extracellular matrix. Moreover, PCs are believed to have mesenchymal stem properties and, therefore, might contribute to regenerative therapy. Here, we review novel ideas concerning PC characteristics and functions in RVDs and discuss potential therapeutic strategies based on PCs, including the targeting of pathological signals and cell-based regenerative treatments.
Collapse
Affiliation(s)
- Quan Zhang
- Department of Ophthalmology, Eye Institute of Chinese PLA, Xijing Hospital, Air Force Medical University, Xi'an, China
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Department of Biochemistry and Molecular Biology, Air Force Medical University, Xi'an, China
| | - Xianchun Yan
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Department of Biochemistry and Molecular Biology, Air Force Medical University, Xi'an, China
| | - Hua Han
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Department of Biochemistry and Molecular Biology, Air Force Medical University, Xi'an, China
| | - Yusheng Wang
- Department of Ophthalmology, Eye Institute of Chinese PLA, Xijing Hospital, Air Force Medical University, Xi'an, China
| | - Jiaxing Sun
- Department of Ophthalmology, Eye Institute of Chinese PLA, Xijing Hospital, Air Force Medical University, Xi'an, China
- Department of Neurobiology, Air Force Medical University, Xi'an, China
| |
Collapse
|
22
|
Lin Y, Gahn J, Banerjee K, Dobreva G, Singhal M, Dubrac A, Ola R. Role of endothelial PDGFB in arterio-venous malformations pathogenesis. Angiogenesis 2024; 27:193-209. [PMID: 38070064 PMCID: PMC11021264 DOI: 10.1007/s10456-023-09900-w] [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: 05/30/2023] [Accepted: 11/05/2023] [Indexed: 04/17/2024]
Abstract
Arterial-venous malformations (AVMs) are direct connections between arteries and veins without an intervening capillary bed. Either familial inherited or sporadically occurring, localized pericytes (PCs) drop is among the AVMs' hallmarks. Whether impaired PC coverage triggers AVMs or it is a secondary event is unclear. Here we evaluated the role of the master regulator of PC recruitment, Platelet derived growth factor B (PDGFB) in AVM pathogenesis. Using tamoxifen-inducible deletion of Pdgfb in endothelial cells (ECs), we show that disruption of EC Pdgfb-mediated PC recruitment and maintenance leads to capillary enlargement and organotypic AVM-like structures. These vascular lesions contain non-proliferative hyperplastic, hypertrophic and miss-oriented capillary ECs with an altered capillary EC fate identity. Mechanistically, we propose that PDGFB maintains capillary EC size and caliber to limit hemodynamic changes, thus restricting expression of Krüppel like factor 4 and activation of Bone morphogenic protein, Transforming growth factor β and NOTCH signaling in ECs. Furthermore, our study emphasizes that inducing or activating PDGFB signaling may be a viable therapeutic approach for treating vascular malformations.
Collapse
Affiliation(s)
- Yanzhu Lin
- Experimental Pharmacology Mannheim (EPM), European Center for Angioscience (ECAS), Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Johannes Gahn
- Experimental Pharmacology Mannheim (EPM), European Center for Angioscience (ECAS), Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Kuheli Banerjee
- Experimental Pharmacology Mannheim (EPM), European Center for Angioscience (ECAS), Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Gergana Dobreva
- Department of Cardiovascular Genomics and Epigenomics, European Center for Angioscience (ECAS), Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
- German Centre for Cardiovascular Research (DZHK), Heidelberg, Germany
| | - Mahak Singhal
- Laboratory of AngioRhythms, European Center for Angioscience (ECAS), Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Alexandre Dubrac
- Centre de Recherche, CHU St. Justine, Montreal, QC, H3T 1C5, Canada
- Département de Pathologie et Biologie Cellulaire, Université de Montréal, Montreal, QC, H3T 1J4, Canada
| | - Roxana Ola
- Experimental Pharmacology Mannheim (EPM), European Center for Angioscience (ECAS), Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany.
| |
Collapse
|
23
|
Hang C, Zu L, Luo X, Wang Y, Yan L, Zhang Z, Le K, Huang Y, Ye L, Ying Y, Chen K, Xu X, Lv Q, Du L. Ddx5 Targeted Epigenetic Modification of Pericytes in Pulmonary Hypertension After Intrauterine Growth Restriction. Am J Respir Cell Mol Biol 2024; 70:400-413. [PMID: 38301267 DOI: 10.1165/rcmb.2023-0244oc] [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: 07/04/2023] [Accepted: 02/01/2024] [Indexed: 02/03/2024] Open
Abstract
Newborns with intrauterine growth restriction (IUGR) have a higher likelihood of developing pulmonary arterial hypertension (PAH) in adulthood. Although there is increasing evidence suggesting that pericytes play a role in regulating myofibroblast transdifferentiation and angiogenesis in malignant and cardiovascular diseases, their involvement in the pathogenesis of IUGR-related pulmonary hypertension and the underlying mechanisms remain incompletely understood. To address this issue, a study was conducted using a Sprague-Dawley rat model of IUGR-related pulmonary hypertension. Our investigation revealed increased proliferation and migration of pulmonary microvascular pericytes in IUGR-related pulmonary hypertension, accompanied by weakened endothelial-pericyte interactions. Through whole-transcriptome sequencing, Ddx5 (DEAD-box protein 5) was identified as one of the hub genes in pericytes. DDX5, a member of the RNA helicase family, plays a role in the regulation of ATP-dependent RNA helicase activities and cellular function. MicroRNAs have been implicated in the pathogenesis of PAH, and microRNA-205 (miR-205) regulates cell proliferation, migration, and angiogenesis. The results of dual-luciferase reporter assays confirmed the specific binding of miR-205 to Ddx5. Mechanistically, miR-205 negatively regulates Ddx5, leading to the degradation of β-catenin by inhibiting the phosphorylation of Gsk3β at serine 9. In vitro experiments showed the addition of miR-205 effectively ameliorated pericyte dysfunction. Furthermore, in vivo experiments demonstrated that miR-205 agomir could ameliorate pulmonary hypertension. Our findings indicated that the downregulation of miR-205 expression mediates pericyte dysfunction through the activation of Ddx5. Therefore, targeting the miR-205/Ddx5/p-Gsk3β/β-catenin axis could be a promising therapeutic approach for IUGR-related pulmonary hypertension.
Collapse
Affiliation(s)
| | - Lu Zu
- Department of Neonatology and
| | - Xiaofei Luo
- Department of Pediatrics, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, People's Republic of China; and
| | - Yu Wang
- Department of Neonatology and
| | - Lingling Yan
- Department of Pediatrics, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, People's Republic of China; and
| | | | - Kaixing Le
- Academy of Pediatrics, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, People's Republic of China
| | | | | | | | | | - Xuefeng Xu
- Department of Rheumatology, Immunology, and Allergy, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, Zhejiang Province, People's Republic of China
| | | | | |
Collapse
|
24
|
Ishiguro H, Ushiki T, Honda A, Yoshimatsu Y, Ohashi R, Okuda S, Kawasaki A, Cho K, Tamura S, Suwabe T, Katagiri T, Ling Y, Iijima A, Mikami T, Kitagawa H, Uemura A, Sango K, Masuko M, Igarashi M, Sone H. Reduced chondroitin sulfate content prevents diabetic neuropathy through transforming growth factor-β signaling suppression. iScience 2024; 27:109528. [PMID: 38595797 PMCID: PMC11002665 DOI: 10.1016/j.isci.2024.109528] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 11/08/2023] [Accepted: 03/15/2024] [Indexed: 04/11/2024] Open
Abstract
Diabetic neuropathy (DN) is a major complication of diabetes mellitus. Chondroitin sulfate (CS) is one of the most important extracellular matrix components and is known to interact with various diffusible factors; however, its role in DN pathology has not been examined. Therefore, we generated CSGalNAc-T1 knockout (T1KO) mice, in which CS levels were reduced. We demonstrated that diabetic T1KO mice were much more resistant to DN than diabetic wild-type (WT) mice. We also found that interactions between pericytes and vascular endothelial cells were more stable in T1KO mice. Among the RNA-seq results, we focused on the transforming growth factor β signaling pathway and found that the phosphorylation of Smad2/3 was less upregulated in T1KO mice than in WT mice under hyperglycemic conditions. Taken together, a reduction in CS level attenuates DN progression, indicating that CS is an important factor in DN pathogenesis.
Collapse
Affiliation(s)
- Hajime Ishiguro
- Departments of Hematology, Endocrinology, and Metabolism, Graduate School of Medical and Dental Sciences, Niigata university, Niigata, Japan
| | - Takashi Ushiki
- Departments of Hematology, Endocrinology, and Metabolism, Graduate School of Medical and Dental Sciences, Niigata university, Niigata, Japan
- Division of Hematology and Oncology, Graduate School of Health Sciences, Niigata University, Niigata, Japan
- Departments of Transfusion Medicine, Cell Therapy and Regenerative Medicine, Medical and Dental Hospital, Niigata University, Niigata, Japan
| | - Atsuko Honda
- Department of Neurochemistry and Molecular Cell Biology, Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Japan
- Center for Research Promotion, Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Japan
| | - Yasuhiro Yoshimatsu
- Division of Pharmacology, Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Japan
| | - Riuko Ohashi
- Divisions of Molecular and Diagnostic Pathology, Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Japan
| | - Shujiro Okuda
- Division of Bioinformatics, Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Asami Kawasaki
- Department of Neurochemistry and Molecular Cell Biology, Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Japan
| | - Kaori Cho
- Departments of Hematology, Endocrinology, and Metabolism, Graduate School of Medical and Dental Sciences, Niigata university, Niigata, Japan
| | - Suguru Tamura
- Departments of Hematology, Endocrinology, and Metabolism, Graduate School of Medical and Dental Sciences, Niigata university, Niigata, Japan
| | - Tatsuya Suwabe
- Departments of Hematology, Endocrinology, and Metabolism, Graduate School of Medical and Dental Sciences, Niigata university, Niigata, Japan
| | - Takayuki Katagiri
- Departments of Hematology, Endocrinology, and Metabolism, Graduate School of Medical and Dental Sciences, Niigata university, Niigata, Japan
| | - Yiwei Ling
- Division of Bioinformatics, Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Atsuhiko Iijima
- Neurophysiology & Biomedical Engineering Lab, Interdisciplinary Program of Biomedical Engineering, Assistive Technology and Art and Sports Sciences, Faculty of Engineering, Niigata University Niigata, Niigata, Japan
| | - Tadahisa Mikami
- Laboratory of Biochemistry, Kobe Pharmaceutical University, Kobe, Japan
| | - Hiroshi Kitagawa
- Laboratory of Biochemistry, Kobe Pharmaceutical University, Kobe, Japan
| | - Akiyoshi Uemura
- Department of Retinal Vascular Biology, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Kazunori Sango
- Diabetic Neuropathy Project, Department of Diseases and Infection, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| | - Masayoshi Masuko
- Departments of Hematology, Endocrinology, and Metabolism, Graduate School of Medical and Dental Sciences, Niigata university, Niigata, Japan
- Hematopoietic Cell Transplantation Niigata University Medical and Dental Hospital, , Niigata University, Niigata, Japan
| | - Michihiro Igarashi
- Department of Neurochemistry and Molecular Cell Biology, Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Japan
| | - Hirohito Sone
- Departments of Hematology, Endocrinology, and Metabolism, Graduate School of Medical and Dental Sciences, Niigata university, Niigata, Japan
| |
Collapse
|
25
|
Carvalho TP, Toledo FAO, Bautista DFA, Silva MF, Oliveira JBS, Lima PA, Costa FB, Ribeiro NQ, Lee JY, Birbrair A, Paixão TA, Tsolis RM, Santos RL. Pericytes modulate endothelial inflammatory response during bacterial infection. mBio 2024; 15:e0325223. [PMID: 38289074 PMCID: PMC10936204 DOI: 10.1128/mbio.03252-23] [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: 12/12/2023] [Accepted: 12/13/2023] [Indexed: 03/14/2024] Open
Abstract
Pericytes are located around blood vessels, in close contact with endothelial cells. We discovered that pericytes dampen pro-inflammatory endothelial cell responses. Endothelial cells co-cultured with pericytes had markedly reduced expression of adhesion molecules (PECAM-1 and ICAM-1) and proinflammatory cytokines (CCL-2 and IL-6) in response to bacterial stimuli (Brucella ovis, Listeria monocytogenes, or Escherichia coli lipopolysaccharide). Pericyte-depleted mice intraperitoneally inoculated with either B. ovis, a stealthy pathogen that does not trigger detectable inflammation, or Listeria monocytogenes, developed peritonitis. Further, during Citrobacter rodentium infection, pericyte-depleted mice developed severe intestinal inflammation, which was not evident in control mice. The anti-inflammatory effect of pericytes required connexin 43, as either chemical inhibition or silencing of connexin 43 abrogated pericyte-mediated suppression of endothelial inflammatory responses. Our results define a mechanism by which pericytes modulate inflammation during infection, which shifts our understanding of pericyte biology: from a structural cell to a pro-active player in modulating inflammation. IMPORTANCE A previously unknown mechanism by which pericytes modulate inflammation was discovered. The absence of pericytes or blocking interaction between pericytes and endothelium through connexin 43 results in stronger inflammation, which shifts our understanding of pericyte biology, from a structural cell to a player in controlling inflammation.
Collapse
Affiliation(s)
- Thaynara P. Carvalho
- Departamento de Clínica e Cirurgia Veterinárias, Escola de Veterinária, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
- Department of Medical Microbiology and Immunology, University of California, Davis, California, USA
| | - Frank A. O. Toledo
- Departamento de Clínica e Cirurgia Veterinárias, Escola de Veterinária, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Diego F. A. Bautista
- Departamento de Clínica e Cirurgia Veterinárias, Escola de Veterinária, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Monique F. Silva
- Departamento de Patologia Geral, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Jefferson B. S. Oliveira
- Departamento de Clínica e Cirurgia Veterinárias, Escola de Veterinária, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Pâmela A. Lima
- Departamento de Clínica e Cirurgia Veterinárias, Escola de Veterinária, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Fabíola B. Costa
- Departamento de Clínica e Cirurgia Veterinárias, Escola de Veterinária, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Noelly Q. Ribeiro
- Departamento de Clínica e Cirurgia Veterinárias, Escola de Veterinária, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Jee-Yon Lee
- Department of Medical Microbiology and Immunology, University of California, Davis, California, USA
| | - Alexander Birbrair
- Departamento de Patologia Geral, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
- Department of Dermatology, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Tatiane A. Paixão
- Departamento de Patologia Geral, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Reneé M. Tsolis
- Department of Medical Microbiology and Immunology, University of California, Davis, California, USA
| | - Renato L. Santos
- Departamento de Clínica e Cirurgia Veterinárias, Escola de Veterinária, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
- Department of Medical Microbiology and Immunology, University of California, Davis, California, USA
| |
Collapse
|
26
|
Van Hulle C, Ince S, Okonkwo OC, Bendlin BB, Johnson SC, Carlsson CM, Asthana S, Love S, Blennow K, Zetterberg H, Scott Miners J. Elevated CSF angiopoietin-2 correlates with blood-brain barrier leakiness and markers of neuronal injury in early Alzheimer's disease. Transl Psychiatry 2024; 14:3. [PMID: 38182581 PMCID: PMC10770135 DOI: 10.1038/s41398-023-02706-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Revised: 11/12/2023] [Accepted: 12/05/2023] [Indexed: 01/07/2024] Open
Abstract
Breakdown of the neurovascular unit is associated with blood-brain barrier (BBB) leakiness contributing to cognitive decline and disease pathology in the early stages of Alzheimer's disease (AD). Vascular stability depends on angiopoietin-1 (ANGPT-1) signalling, antagonised by angiopoietin-2 (ANGPT-2) expressed upon endothelial injury. We examined the relationship between CSF ANGPT-2 and CSF markers of BBB leakiness and core AD biomarkers across three independent cohorts: (i) 31 AD patients and 33 healthy controls grouped according to their biomarker profile (i.e., AD cases t-tau > 400 pg/mL, p-tau > 60 pg/mL and Aβ42 < 550 pg/mL); (ii) 121 participants in the Wisconsin Registry for Alzheimer's Prevention or Wisconsin Alzheimer's Disease Research study (84 participants cognitively unimpaired (CU) enriched for a parental history of AD, 20 participants with mild cognitive impairment (MCI), and 17 with AD); (iii) a neurologically normal cohort aged 23-78 years with paired CSF and serum samples. CSF ANGPT-2, sPDGFRβ, albumin and fibrinogen levels were measured by sandwich ELISA. In cohort (i), CSF ANGPT-2 was elevated in AD and correlated with CSF t-tau and p-tau181 but not Aβ42. ANGPT-2 also correlated positively with CSF sPDGFRβ and fibrinogen - markers of pericyte injury and BBB leakiness. In cohort (ii), CSF ANGPT-2 was highest in MCI and correlated with CSF albumin in the CU and MCI cohorts but not in AD. CSF ANGPT-2 also correlated with CSF t-tau and p-tau and with markers of neuronal injury (neurogranin and α-synuclein) and neuroinflammation (GFAP and YKL-40). In cohort (iii), CSF ANGPT-2 correlated strongly with the CSF/serum albumin ratio. Serum ANGPT-2 showed non-significant positive associations with CSF ANGPT-2 and the CSF/serum albumin ratio. Together, these data indicate that CSF and possibly serum ANGPT-2 is associated with BBB leakiness in early AD and is closely related to tau pathology and neuronal injury. The utility of serum ANGPT-2 as a biomarker of BBB damage in AD requires further study.
Collapse
Affiliation(s)
- Carol Van Hulle
- Wisconsin Alzheimer's Disease Research Center, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, USA
- Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, University of Wisconsin-Madison, Madison, WI, USA
| | - Selvi Ince
- Dementia Research Group, Clinical Neurosciences, Bristol Medical School, University of Bristol, Bristol, UK
| | - Ozioma C Okonkwo
- Wisconsin Alzheimer's Disease Research Center, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, USA
- Wisconsin Alzheimer's Institute, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, USA
- Geriatric Research Education and Clinical Center, William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
| | - Barbara B Bendlin
- Wisconsin Alzheimer's Disease Research Center, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, USA
- Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, University of Wisconsin-Madison, Madison, WI, USA
- Wisconsin Alzheimer's Institute, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, USA
- Geriatric Research Education and Clinical Center, William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
| | - Sterling C Johnson
- Wisconsin Alzheimer's Disease Research Center, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, USA
- Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, University of Wisconsin-Madison, Madison, WI, USA
- Wisconsin Alzheimer's Institute, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, USA
- Geriatric Research Education and Clinical Center, William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
| | - Cynthia M Carlsson
- Wisconsin Alzheimer's Disease Research Center, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, USA
- Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, University of Wisconsin-Madison, Madison, WI, USA
- Wisconsin Alzheimer's Institute, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, USA
- Geriatric Research Education and Clinical Center, William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
| | - Sanjay Asthana
- Wisconsin Alzheimer's Disease Research Center, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, USA
- Wisconsin Alzheimer's Institute, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, USA
| | - Seth Love
- Dementia Research Group, Clinical Neurosciences, Bristol Medical School, University of Bristol, Bristol, UK
| | - Kaj Blennow
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
| | - Henrik Zetterberg
- Wisconsin Alzheimer's Disease Research Center, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, USA
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
- Department of Neurodegenerative Disease, UCL Institute of Neurology, Queen Square, London, UK
- UK Dementia Research Institute at UCL, London, UK
- Hong Kong Center for Neurodegenerative Diseases, Clear Water Bay, Hong Kong, China
| | - J Scott Miners
- Dementia Research Group, Clinical Neurosciences, Bristol Medical School, University of Bristol, Bristol, UK.
| |
Collapse
|
27
|
van Splunder H, Villacampa P, Martínez-Romero A, Graupera M. Pericytes in the disease spotlight. Trends Cell Biol 2024; 34:58-71. [PMID: 37474376 PMCID: PMC10777571 DOI: 10.1016/j.tcb.2023.06.001] [Citation(s) in RCA: 43] [Impact Index Per Article: 43.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Revised: 06/01/2023] [Accepted: 06/09/2023] [Indexed: 07/22/2023]
Abstract
Pericytes are known as the mural cells in small-caliber vessels that interact closely with the endothelium. Pericytes play a key role in vasculature formation and homeostasis, and when dysfunctional contribute to vasculature-related diseases such as diabetic retinopathy and neurodegenerative conditions. In addition, significant extravascular roles of pathological pericytes are being discovered with relevant implications for cancer and fibrosis. Pericyte research is challenged by the lack of consistent molecular markers and clear discrimination criteria versus other (mural) cells. However, advances in single-cell approaches are uncovering and clarifying mural cell identities, biological functions, and ontogeny across organs. We discuss the latest developments in pericyte pathobiology to inform future research directions and potential outcomes.
Collapse
Affiliation(s)
- Hielke van Splunder
- Endothelial Pathobiology and Microenviroment Group, Josep Carreras Leukemia Research Institute (IJC), 08916 Badalona, Barcelona, Catalonia, Spain
| | - Pilar Villacampa
- Department of Physiological Sciences, Faculty of Medicine and Health Sciences, University of Barcelona and Bellvitge Biomedical Research Institute (IDIBELL), Carrer de la Feixa Llarga s/n, 08907 l'Hospitalet de Llobregat, Barcelona, Spain
| | - Anabel Martínez-Romero
- Endothelial Pathobiology and Microenviroment Group, Josep Carreras Leukemia Research Institute (IJC), 08916 Badalona, Barcelona, Catalonia, Spain
| | - Mariona Graupera
- Endothelial Pathobiology and Microenviroment Group, Josep Carreras Leukemia Research Institute (IJC), 08916 Badalona, Barcelona, Catalonia, Spain; Institución Catalana de Investigación y Estudios Avanzados (ICREA), Passeig de Lluís Companys 23, Barcelona, Spain; Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Instituto de Salud Carlos III, Avenida de Monforte de Lemos 5, 28029 Madrid, Spain.
| |
Collapse
|
28
|
de Gea P, Benkeder S, Bouvet P, Aimard M, Chounlamountri N, Honnorat J, Do LD, Meissirel C. VEGF controls microglial phagocytic response to amyloid-β. Front Cell Neurosci 2023; 17:1264402. [PMID: 38162003 PMCID: PMC10757340 DOI: 10.3389/fncel.2023.1264402] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Accepted: 11/23/2023] [Indexed: 01/03/2024] Open
Abstract
Microglial cells are well known to be implicated in the pathogenesis of Alzheimer's disease (AD), due to the impaired clearance of amyloid-β (Aβ) protein. In AD, Aβ accumulates in the brain parenchyma as soluble oligomers and protofibrils, and its aggregation process further give rise to amyloid plaques. Compelling evidence now indicate that Aβ oligomers (Aβo) are the most toxic forms responsible for neuronal and synaptic alterations. Recently, we showed that the Vascular Endothelial Growth Factor (VEGF) counteracts Aβo-induced synaptic alterations and that a peptide derived from VEGF is able to inhibit Aβ aggregation process. Moreover, VEGF has been reported to promote microglial chemotaxis to Aβ brain deposits. We therefore investigated whether VEGF could influence microglial phagocytic response to Aβ, using in vitro and ex vivo models of amyloid accumulation. We report here that VEGF increases Aβo phagocytosis by microglial cells and further characterized the molecular basis of the VEGF effect. VEGF is able to control α-secretase activity in microglial cells, resulting in the increased cleavage of the Triggering Receptor Expressed on Myeloid cells 2 (TREM2), a major microglial Aβ receptor. Consistently, the soluble form sTREM2 also increases Aβo phagocytosis by microglial cells. Taken together, these findings propose VEGF as a new regulator of Aβ clearance and suggest its potential role in rescuing compromised microglial function in AD.
Collapse
Affiliation(s)
- Priscille de Gea
- Laboratory MeLIS, Institut Neuromyogène, Synaptopathies and Autoantibodies, INSERM U1314, CNRS UMR 5284, Université Claude Bernard Lyon 1, Lyon, France
| | - Sarah Benkeder
- Laboratory MeLIS, Institut Neuromyogène, Synaptopathies and Autoantibodies, INSERM U1314, CNRS UMR 5284, Université Claude Bernard Lyon 1, Lyon, France
| | - Pauline Bouvet
- Laboratory MeLIS, Institut Neuromyogène, Synaptopathies and Autoantibodies, INSERM U1314, CNRS UMR 5284, Université Claude Bernard Lyon 1, Lyon, France
| | - Mélanie Aimard
- Laboratory MeLIS, Institut Neuromyogène, Synaptopathies and Autoantibodies, INSERM U1314, CNRS UMR 5284, Université Claude Bernard Lyon 1, Lyon, France
| | - Naura Chounlamountri
- Laboratory MeLIS, Institut Neuromyogène, Synaptopathies and Autoantibodies, INSERM U1314, CNRS UMR 5284, Université Claude Bernard Lyon 1, Lyon, France
| | - Jérôme Honnorat
- Laboratory MeLIS, Institut Neuromyogène, Synaptopathies and Autoantibodies, INSERM U1314, CNRS UMR 5284, Université Claude Bernard Lyon 1, Lyon, France
- French Reference Center for Paraneoplastic Neurological Syndromes and Autoimmune Encephalitis, Hospices Civils de Lyon, Hôpital Neurologique Pierre Wertheimer, Bron, France
| | - Le Duy Do
- Laboratory MeLIS, Institut Neuromyogène, Synaptopathies and Autoantibodies, INSERM U1314, CNRS UMR 5284, Université Claude Bernard Lyon 1, Lyon, France
- French Reference Center for Paraneoplastic Neurological Syndromes and Autoimmune Encephalitis, Hospices Civils de Lyon, Hôpital Neurologique Pierre Wertheimer, Bron, France
| | - Claire Meissirel
- Laboratory MeLIS, Institut Neuromyogène, Synaptopathies and Autoantibodies, INSERM U1314, CNRS UMR 5284, Université Claude Bernard Lyon 1, Lyon, France
| |
Collapse
|
29
|
Teske NC, Dyckhoff-Shen S, Beckenbauer P, Bewersdorf JP, Engelen-Lee JY, Hammerschmidt S, Kälin RE, Pfister HW, Brouwer MC, Klein M, Glass R, van de Beek D, Koedel U. Pericytes are protective in experimental pneumococcal meningitis through regulating leukocyte infiltration and blood-brain barrier function. J Neuroinflammation 2023; 20:267. [PMID: 37978545 PMCID: PMC10655320 DOI: 10.1186/s12974-023-02938-z] [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: 07/16/2023] [Accepted: 10/27/2023] [Indexed: 11/19/2023] Open
Abstract
BACKGROUND Brain pericytes participate in the regulation of cerebral blood flow and the maintenance of blood-brain barrier integrity. Because of their perivascular localization, their receptor repertoire, and their potential ability to respond to inflammatory and infectious stimuli by producing various cytokines and chemokines, these cells are also thought to play an active role in the immune response to brain infections. This assumption is mainly supported by in vitro studies, investigations in in vivo disease models are largely missing. Here, we analysed the role of brain pericytes in pneumococcal meningitis, in vitro and in vivo in two animal models of pneumococcal meningitis. METHODS Primary murine and human pericytes were stimulated with increasing concentrations of different serotypes of Streptococcus pneumoniae in the presence or absence of Toll-like receptor inhibitors and their cell viability and cytokine production were monitored. To gain insight into the role of pericytes in brain infection in vivo, we performed studies in a zebrafish embryo model of pneumococcal meningitis in which pericytes were pharmacologically depleted. Furthermore, we analyzed the impact of genetically induced pericyte ablation on disease progression, intracranial complications, and brain inflammation in an adult mouse model of this disease. RESULTS Both murine and human pericytes reacted to pneumococcal exposure with the release of selected cytokines. This cytokine release is pneumolysin-dependent, TLR-dependent in murine (but not human) pericytes and can be significantly increased by macrophage-derived IL-1b. Pharmacological depletion of pericytes in zebrafish embryos resulted in increased cerebral edema and mortality due to pneumococcal meningitis. Correspondingly, in an adult mouse meningitis model, a more pronounced blood-brain barrier disruption and leukocyte infiltration, resulting in an unfavorable disease course, was observed following genetic pericyte ablation. The degree of leukocyte infiltration positively correlated with an upregulation of chemokine expression in the brains of pericyte-depleted mice. CONCLUSIONS Our findings show that pericytes play a protective role in pneumococcal meningitis by impeding leukocyte migration and preventing blood-brain barrier breaching. Thus, preserving the integrity of the pericyte population has the potential as a new therapeutic strategy in pneumococcal meningitis.
Collapse
Affiliation(s)
- Nina C Teske
- Department of Neurology, LMU University Hospital, LMU Munich, Munich, Germany.
- ESCMID Study Group for Infections of the Brain, Basel, Switzerland.
- Department of Neurology, Amsterdam UMC, University of Amsterdam, Amsterdam Neuroscience, Amsterdam, The Netherlands.
- Department of Neurosurgery, LMU University Hospital, LMU Munich, Munich, Germany.
| | | | - Paul Beckenbauer
- Department of Neurology, LMU University Hospital, LMU Munich, Munich, Germany
| | | | - Joo-Yeon Engelen-Lee
- ESCMID Study Group for Infections of the Brain, Basel, Switzerland
- Department of Neurology, Amsterdam UMC, University of Amsterdam, Amsterdam Neuroscience, Amsterdam, The Netherlands
| | - Sven Hammerschmidt
- Department Genetics of Microorganisms, Interfaculty Institute of Genetics and Functional Genomics, University of Greifswald, Greifswald, Germany
| | - Roland E Kälin
- Neurosurgical Research, Department of Neurosurgery, LMU University Hospital, LMU Munich, Munich, Germany
- Walter Brendel Center of Experimental Medicine, Faculty of Medicine, LMU Munich, Munich, Germany
| | - Hans-Walter Pfister
- Department of Neurology, LMU University Hospital, LMU Munich, Munich, Germany
- ESCMID Study Group for Infections of the Brain, Basel, Switzerland
| | - Matthijs C Brouwer
- ESCMID Study Group for Infections of the Brain, Basel, Switzerland
- Department of Neurology, Amsterdam UMC, University of Amsterdam, Amsterdam Neuroscience, Amsterdam, The Netherlands
| | - Matthias Klein
- Department of Neurology, LMU University Hospital, LMU Munich, Munich, Germany
- ESCMID Study Group for Infections of the Brain, Basel, Switzerland
| | - Rainer Glass
- Neurosurgical Research, Department of Neurosurgery, LMU University Hospital, LMU Munich, Munich, Germany
| | - Diederik van de Beek
- ESCMID Study Group for Infections of the Brain, Basel, Switzerland
- Department of Neurology, Amsterdam UMC, University of Amsterdam, Amsterdam Neuroscience, Amsterdam, The Netherlands
| | - Uwe Koedel
- Department of Neurology, LMU University Hospital, LMU Munich, Munich, Germany
- ESCMID Study Group for Infections of the Brain, Basel, Switzerland
| |
Collapse
|
30
|
Lincke JB, Christe L, Unterlauft JD, Zinkernagel MS, Zysset-Burri DC. Microbiome and Retinal Vascular Diseases. THE AMERICAN JOURNAL OF PATHOLOGY 2023; 193:1675-1682. [PMID: 36963629 DOI: 10.1016/j.ajpath.2023.02.017] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/06/2022] [Revised: 02/06/2023] [Accepted: 02/16/2023] [Indexed: 03/26/2023]
Abstract
The gut microbiome consists of more than a thousand different microbes and their associated genes and microbial metabolites. It influences various host metabolic pathways and is therefore important for homeostasis. In recent years, its influence on health and disease has been extensively researched. Dysbiosis, or imbalance in the gut microbiome, is associated with several diseases. Consequent chronic inflammation may lead to or promote inflammatory bowel disease, obesity, diabetes mellitus, atherosclerosis, alcoholic and non-alcoholic liver disease, cirrhosis, hepatocellular carcinoma, and other diseases. The pathogenesis of the three most common retinal vascular diseases, diabetic retinopathy, retinal vein occlusion, and retinal artery occlusion, may also be influenced by an altered microbiome and associated risk factors such as diabetes mellitus, atherosclerosis, hypertension, and obesity. Direct cause-effect relationships remain less well understood. A potential prevention or treatment modality for these diseases could be targeting and modulating the individual's gut microbiome.
Collapse
Affiliation(s)
- Joel-Benjamin Lincke
- Department of Ophthalmology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland.
| | - Lucine Christe
- Institute of Pathology, University of Bern, Bern, Switzerland
| | - Jan Darius Unterlauft
- Department of Ophthalmology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Martin S Zinkernagel
- Department of Ophthalmology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland; Department of BioMedical Research, University of Bern, Bern, Switzerland
| | - Denise C Zysset-Burri
- Department of Ophthalmology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland; Department of BioMedical Research, University of Bern, Bern, Switzerland
| |
Collapse
|
31
|
Alarcon-Martinez L, Shiga Y, Villafranca-Baughman D, Cueva Vargas JL, Vidal Paredes IA, Quintero H, Fortune B, Danesh-Meyer H, Di Polo A. Neurovascular dysfunction in glaucoma. Prog Retin Eye Res 2023; 97:101217. [PMID: 37778617 DOI: 10.1016/j.preteyeres.2023.101217] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Revised: 09/23/2023] [Accepted: 09/25/2023] [Indexed: 10/03/2023]
Abstract
Retinal ganglion cells, the neurons that die in glaucoma, are endowed with a high metabolism requiring optimal provision of oxygen and nutrients to sustain their activity. The timely regulation of blood flow is, therefore, essential to supply firing neurons in active areas with the oxygen and glucose they need for energy. Many glaucoma patients suffer from vascular deficits including reduced blood flow, impaired autoregulation, neurovascular coupling dysfunction, and blood-retina/brain-barrier breakdown. These processes are tightly regulated by a community of cells known as the neurovascular unit comprising neurons, endothelial cells, pericytes, Müller cells, astrocytes, and microglia. In this review, the neurovascular unit takes center stage as we examine the ability of its members to regulate neurovascular interactions and how their function might be altered during glaucomatous stress. Pericytes receive special attention based on recent data demonstrating their key role in the regulation of neurovascular coupling in physiological and pathological conditions. Of particular interest is the discovery and characterization of tunneling nanotubes, thin actin-based conduits that connect distal pericytes, which play essential roles in the complex spatial and temporal distribution of blood within the retinal capillary network. We discuss cellular and molecular mechanisms of neurovascular interactions and their pathophysiological implications, while highlighting opportunities to develop strategies for vascular protection and regeneration to improve functional outcomes in glaucoma.
Collapse
Affiliation(s)
- Luis Alarcon-Martinez
- Department of Neuroscience, Université de Montréal, PO Box 6128, Station centre-ville, Montreal, QC, Canada; Neuroscience Division, Centre de recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), 900 Saint Denis Street, Montreal, QC, Canada; Centre for Eye Research Australia, University of Melbourne, Melbourne, Australia
| | - Yukihiro Shiga
- Department of Neuroscience, Université de Montréal, PO Box 6128, Station centre-ville, Montreal, QC, Canada; Neuroscience Division, Centre de recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), 900 Saint Denis Street, Montreal, QC, Canada
| | - Deborah Villafranca-Baughman
- Department of Neuroscience, Université de Montréal, PO Box 6128, Station centre-ville, Montreal, QC, Canada; Neuroscience Division, Centre de recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), 900 Saint Denis Street, Montreal, QC, Canada
| | - Jorge L Cueva Vargas
- Department of Neuroscience, Université de Montréal, PO Box 6128, Station centre-ville, Montreal, QC, Canada; Neuroscience Division, Centre de recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), 900 Saint Denis Street, Montreal, QC, Canada
| | - Isaac A Vidal Paredes
- Department of Neuroscience, Université de Montréal, PO Box 6128, Station centre-ville, Montreal, QC, Canada; Neuroscience Division, Centre de recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), 900 Saint Denis Street, Montreal, QC, Canada
| | - Heberto Quintero
- Department of Neuroscience, Université de Montréal, PO Box 6128, Station centre-ville, Montreal, QC, Canada; Neuroscience Division, Centre de recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), 900 Saint Denis Street, Montreal, QC, Canada
| | - Brad Fortune
- Discoveries in Sight Research Laboratories, Devers Eye Institute and Legacy Research Institute, Legacy Healthy, Portland, OR, USA
| | - Helen Danesh-Meyer
- Department of Ophthalmology, New Zealand National Eye Centre, Faculty of Medical and Health Sciences, University of Auckland, New Zealand
| | - Adriana Di Polo
- Department of Neuroscience, Université de Montréal, PO Box 6128, Station centre-ville, Montreal, QC, Canada; Neuroscience Division, Centre de recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), 900 Saint Denis Street, Montreal, QC, Canada.
| |
Collapse
|
32
|
Takamura Y, Yamada Y, Morioka M, Gozawa M, Matsumura T, Inatani M. Turnover of Microaneurysms After Intravitreal Injections of Faricimab for Diabetic Macular Edema. Invest Ophthalmol Vis Sci 2023; 64:31. [PMID: 37856112 PMCID: PMC10593137 DOI: 10.1167/iovs.64.13.31] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2023] [Accepted: 10/05/2023] [Indexed: 10/20/2023] Open
Abstract
Purpose Microaneurysm (MA) plays an important role in the pathogenesis of diabetic macular edema (DME) progression and response to anti-vascular endothelial growth factor (VEGF) therapy. This study aimed to investigate the effect of faricimab, a bispecific antibody against angiopoietin-2 and VEGF, on the number of MAs and their turnover in the treatment of DME. Methods We included that patients with DME who underwent three monthly injections of faricimab in one eye, with the other eye as control. We examined central retinal thickness (CRT) based on optical coherence tomography (OCT) and best-corrected visual acuity. Turnover, including loss and newly formed MAs, and the total number of MAs were counted based on merged images of the OCT map and fluorescein angiography. Results We enrolled 28 patients with DME. After 3 monthly injections of faricimab, CRT significantly improved, 66.0 ± 16.2% of MAs disappeared, and 6.71 ± 5.6% of new MAs were generated, resulting in total reduction to 40.7 ± 15.2%. In the treated eyes, MA disappearance (P < 0.0001) and turnover (P = 0.007) were significantly greater, and new formation was smaller (P < 0.0001) than in non-treated eyes. The size of the retained MAs decreased after treatment. Microaneurysm turnover was not significantly different between areas with and without edema before treatment. Conclusions In the process of improving edema in DME with faricimab, MAs shrink and disappear, and formation of MAs are inhibited, resulting in decreased total number of MAs. Intravitreal administration of faricimab suppresses vascular permeability and improves vascular structure.
Collapse
Affiliation(s)
- Yoshihiro Takamura
- Department of Ophthalmology, Faculty of Medical Sciences, University of Fukui, Eiheiji-cho, Yoshida-gun, Fukui-ken, Japan
| | - Yutaka Yamada
- Department of Ophthalmology, Faculty of Medical Sciences, University of Fukui, Eiheiji-cho, Yoshida-gun, Fukui-ken, Japan
| | - Masakazu Morioka
- Department of Ophthalmology, Faculty of Medical Sciences, University of Fukui, Eiheiji-cho, Yoshida-gun, Fukui-ken, Japan
| | - Makoto Gozawa
- Department of Ophthalmology, Faculty of Medical Sciences, University of Fukui, Eiheiji-cho, Yoshida-gun, Fukui-ken, Japan
| | - Takehiro Matsumura
- Department of Ophthalmology, Faculty of Medical Sciences, University of Fukui, Eiheiji-cho, Yoshida-gun, Fukui-ken, Japan
| | - Masaru Inatani
- Department of Ophthalmology, Faculty of Medical Sciences, University of Fukui, Eiheiji-cho, Yoshida-gun, Fukui-ken, Japan
| |
Collapse
|
33
|
Horikami D, Sekihachi E, Omori K, Kobayashi Y, Kobayashi K, Nagata N, Kurata K, Uemura A, Murata T. Roles of lipocalin-type and hematopoietic prostaglandin D synthases in mouse retinal angiogenesis. J Lipid Res 2023; 64:100439. [PMID: 37666361 PMCID: PMC10571029 DOI: 10.1016/j.jlr.2023.100439] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Revised: 08/19/2023] [Accepted: 08/22/2023] [Indexed: 09/06/2023] Open
Abstract
Normal angiogenesis is essential for retinal development and maintenance of visual function in the eye, and its abnormality can cause retinopathy and other eye diseases. Prostaglandin D2 is an anti-angiogenic lipid mediator produced by lipocalin-type PGD synthase (L-PGDS) or hematopoietic PGD synthase (H-PGDS). However, the exact role of these PGD synthases remains unclear. Therefore, we compared the roles of these synthases in murine retinal angiogenesis under physiological and pathological conditions. On postnatal day (P) 8, the WT murine retina was covered with an elongated vessel. L-PGDS deficiency, but not H-PGDS, reduced the physiological vessel elongation with sprouts increase. L-PGDS expression was observed in endothelial cells and neural cells. In vitro, L-PGDS inhibition increased the hypoxia-induced vascular endothelial growth factor expression in isolated endothelial cells, inhibited by a prostaglandin D2 metabolite, 15-deoxy-Δ12,14 -PGJ2 (15d-PGJ2) treatment. Pericyte depletion, using antiplatelet-derived growth factor receptor-β antibody, caused retinal hemorrhage with vessel elongation impairment and macrophage infiltration in the WT P8 retina. H-PGDS deficiency promoted hemorrhage but inhibited the impairment of vessel elongation, while L-PGDS did not. In the pericyte-depleted WT retina, H-PGDS was expressed in the infiltrated macrophages. Deficiency of the D prostanoid receptor also inhibited the vessel elongation impairment. These results suggest the endogenous role of L-PGDS signaling in physiological angiogenesis and that of H-PGDS/D prostanoid 1 signaling in pathological angiogenesis.
Collapse
Affiliation(s)
- Daiki Horikami
- Department of Animal Radiology, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Bunkyo-ku, Tokyo, Japan
| | - Erika Sekihachi
- Department of Animal Radiology, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Bunkyo-ku, Tokyo, Japan
| | - Keisuke Omori
- Department of Animal Radiology, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Bunkyo-ku, Tokyo, Japan
| | - Yui Kobayashi
- Department of Animal Radiology, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Bunkyo-ku, Tokyo, Japan
| | - Koji Kobayashi
- Department of Animal Radiology, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Bunkyo-ku, Tokyo, Japan
| | - Nanae Nagata
- Department of Animal Radiology, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Bunkyo-ku, Tokyo, Japan
| | - Kaori Kurata
- Department of Retinal Vascular Biology, Nagoya City University Graduate School of Medical Sciences, Mizuho-ku, Nagoya, Japan
| | - Akiyoshi Uemura
- Department of Retinal Vascular Biology, Nagoya City University Graduate School of Medical Sciences, Mizuho-ku, Nagoya, Japan
| | - Takahisa Murata
- Department of Animal Radiology, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Bunkyo-ku, Tokyo, Japan.
| |
Collapse
|
34
|
Mangalesh S, Toth CA. Preterm infant retinal OCT markers of perinatal health and retinopathy of prematurity. Front Pediatr 2023; 11:1238193. [PMID: 37808559 PMCID: PMC10551634 DOI: 10.3389/fped.2023.1238193] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/11/2023] [Accepted: 08/29/2023] [Indexed: 10/10/2023] Open
Abstract
The increasing survival of preterm infants has led to the importance of improving long-term outcomes associated with preterm birth. Antenatal and perinatal insults not only impact mortality, but also long-term disability. While in the intensive care nursery, preterm infants are also exposed to various stressors that lead to long-term cognitive deficits. It is therefore critical to identify early, low-stress, non-invasive biomarkers for preterm infant health. Optical coherence tomography (OCT) is a powerful imaging modality that has recently been adapted to the infant population and provides noninvasive, high-resolution, cross-sectional imaging of the infant eye at the bedside with low stress relative to conventional examination. In this review we delve into discussing the associations between preterm systemic health factors and OCT-based retinal findings and their potential contribution to the development of non-invasive biomarkers for infant health and for retinopathy of prematurity (ROP).
Collapse
Affiliation(s)
| | - Cynthia A. Toth
- Department of Ophthalmology, Duke University School of Medicine, Durham, NC, United States
| |
Collapse
|
35
|
Tatsukawa T, Kano K, Nakajima KI, Yazawa T, Eguchi R, Kabara M, Horiuchi K, Hayasaka T, Matsuo R, Hasebe N, Azuma N, Kawabe JI. NG2-positive pericytes regulate homeostatic maintenance of slow-type skeletal muscle with rapid myonuclear turnover. Stem Cell Res Ther 2023; 14:205. [PMID: 37592340 PMCID: PMC10433572 DOI: 10.1186/s13287-023-03433-1] [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: 01/24/2023] [Accepted: 07/26/2023] [Indexed: 08/19/2023] Open
Abstract
BACKGROUND Skeletal muscle comprises almost 40% of the human body and is essential for movement, structural support and metabolic homeostasis. Size of multinuclear skeletal muscle is stably maintained under steady conditions with the sporadic fusion of newly produced myocytes to compensate for the muscular turnover caused by daily wear and tear. It is becoming clear that microvascular pericytes (PCs) exhibit myogenic activity. However, whether PCs act as myogenic stem cells for the homeostatic maintenance of skeletal muscles during adulthood remains uncertain. METHODS We utilized PC-fused myofibers using PC-specific lineage tracing mouse (NG2-CreERT/Rosa-tdTomato) to observe whether muscle resident PCs have myogenic potential during daily life. Genetic PC deletion mouse model (NG2-CreERT/DTA) was used to test whether PC differentiates to myofibers for maintenance of muscle structure and function under homeostatic condition. RESULTS Under steady breeding conditions, tdTomato-expressing PCs were infused into myofibers, and subsequently, PC-derived nuclei were incorporated into myofibers. Especially in type-I slow-type myofibers such as the soleus, tdTomato+ myofibers were already observed 3 days after PC labeling; their ratio reached a peak (approximately 80%) within 1 month and was maintained for more than 1 year. Consistently, the NG2+ PC-specific deletion induced muscular atrophy in a slow-type myofiber-specific manner under steady breeding conditions. The number of myonucleus per volume of each myofiber was constant during observation period. CONCLUSIONS These findings demonstrate that the turnover of myonuclei in slow-type myofibers is relatively fast, with PCs acting as myogenic stem cells-the suppliers of new myonuclei under steady conditions-and play a vital role in the homeostatic maintenance of slow-type muscles.
Collapse
Affiliation(s)
- Takamitsu Tatsukawa
- Department of Biochemistry, Asahikawa Medical University, 2-1-1 Midorigaoka-Higashi, Asahikawa, 078-8510, Japan
- Department of Vascular Surgery, Asahikawa Medical University, 2-1-1 Midorigaoka-Higashi, Asahikawa, 078-8510, Japan
| | - Kohei Kano
- Department of Biochemistry, Asahikawa Medical University, 2-1-1 Midorigaoka-Higashi, Asahikawa, 078-8510, Japan
- Department of Cardiovascular Regeneration and Innovation, Asahikawa Medical University, 2-1-1 Midorigaoka-Higashi, Asahikawa, 078-8510, Japan
| | - Kei-Ichi Nakajima
- Department of Biochemistry, Asahikawa Medical University, 2-1-1 Midorigaoka-Higashi, Asahikawa, 078-8510, Japan
| | - Takashi Yazawa
- Department of Biochemistry, Asahikawa Medical University, 2-1-1 Midorigaoka-Higashi, Asahikawa, 078-8510, Japan
| | - Ryoji Eguchi
- Department of Biochemistry, Asahikawa Medical University, 2-1-1 Midorigaoka-Higashi, Asahikawa, 078-8510, Japan
| | - Maki Kabara
- Department of Biochemistry, Asahikawa Medical University, 2-1-1 Midorigaoka-Higashi, Asahikawa, 078-8510, Japan
| | - Kiwamu Horiuchi
- Department of Biochemistry, Asahikawa Medical University, 2-1-1 Midorigaoka-Higashi, Asahikawa, 078-8510, Japan
- Division of Cardiovascular, Respiratory and Neurology, Department of Medicine, Asahikawa Medical University, 2-1-1 Midorigaoka-Higashi, Asahikawa, 078-8510, Japan
| | - Taiki Hayasaka
- Department of Biochemistry, Asahikawa Medical University, 2-1-1 Midorigaoka-Higashi, Asahikawa, 078-8510, Japan
- Division of Cardiovascular, Respiratory and Neurology, Department of Medicine, Asahikawa Medical University, 2-1-1 Midorigaoka-Higashi, Asahikawa, 078-8510, Japan
| | - Risa Matsuo
- Department of Biochemistry, Asahikawa Medical University, 2-1-1 Midorigaoka-Higashi, Asahikawa, 078-8510, Japan
- Department of Dermatology, Asahikawa Medical University, 2-1-1 Midorigaoka-Higashi, Asahikawa, 078-8510, Japan
| | - Naoyuki Hasebe
- Department of Cardiovascular Regeneration and Innovation, Asahikawa Medical University, 2-1-1 Midorigaoka-Higashi, Asahikawa, 078-8510, Japan
- Division of Cardiovascular, Respiratory and Neurology, Department of Medicine, Asahikawa Medical University, 2-1-1 Midorigaoka-Higashi, Asahikawa, 078-8510, Japan
| | - Nobuyoshi Azuma
- Department of Vascular Surgery, Asahikawa Medical University, 2-1-1 Midorigaoka-Higashi, Asahikawa, 078-8510, Japan
| | - Jun-Ichi Kawabe
- Department of Biochemistry, Asahikawa Medical University, 2-1-1 Midorigaoka-Higashi, Asahikawa, 078-8510, Japan.
- Department of Cardiovascular Regeneration and Innovation, Asahikawa Medical University, 2-1-1 Midorigaoka-Higashi, Asahikawa, 078-8510, Japan.
| |
Collapse
|
36
|
Kato F, Nozaki M, Kato A, Yasukawa T. Retinal Microvascular Changes after Intravitreal Triamcinolone Acetonide in Diabetic Macular Edema. J Clin Med 2023; 12:jcm12103475. [PMID: 37240582 DOI: 10.3390/jcm12103475] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Revised: 04/30/2023] [Accepted: 05/10/2023] [Indexed: 05/28/2023] Open
Abstract
Intravitreal injection of triamcinolone acetonide (TA) is essential for clinical treatment in patients who insufficiently respond to vascular endothelial factor inhibitors for diabetic macular edema (DME). The aim of this study was to investigate microvascular changes treated with TA using optical coherence tomography angiography (OCTA). After TA in twelve eyes of eleven patients with central retinal thickness (CRT), there was a 20% or more reduction observed. Visual acuity, the number of microaneurysms, vessel density, and the foveal avascular zone (FAZ) area were compared before and at 2 months after TA. At baseline, the number of microaneurysms was 2.1 ± 1.1 in the superficial capillary plexuses (SCP) and 2.0 ± 1.1 in the deep capillary plexuses (DCP), with a significant decrease post-treatment to 1.0 ± 1.0 for SCP and 0.8 ± 0.8 for DCP (SCP; p = 0.018, DCP; p = 0.008). There was significant enlargement of the FAZ area from 0.28 ± 0.11 mm2 to 0.32 ± 0.14 mm2 (p = 0.041). There was no significant difference in the visual acuity and vessel density of SCP and DCP. Results indicated that OCTA was useful for the evaluation of qualitative and morphological retinal microcirculation and that intravitreal TA may decrease microaneurysms.
Collapse
Affiliation(s)
- Fusae Kato
- Department of Ophthalmology, Toyota Kosei Hospital, Toyota 470-0396, Japan
- Department of Ophthalmology and Visual Science, Nagoya City University Graduate School of Medical Sciences, Nagoya 467-8601, Japan
| | - Miho Nozaki
- Department of Ophthalmology and Visual Science, Nagoya City University Graduate School of Medical Sciences, Nagoya 467-8601, Japan
- Department of Ophthalmology, Nagoya City University East Medical Center, Nagoya 464-8547, Japan
| | - Aki Kato
- Department of Ophthalmology and Visual Science, Nagoya City University Graduate School of Medical Sciences, Nagoya 467-8601, Japan
| | - Tsutomu Yasukawa
- Department of Ophthalmology and Visual Science, Nagoya City University Graduate School of Medical Sciences, Nagoya 467-8601, Japan
| |
Collapse
|
37
|
Li J, Chen C, Zhang L, Ren Y, Li H. PDGFRB upregulation contributes to retinal damages in the rat model of retinal ischemia-reperfusion. Biochem Biophys Res Commun 2023; 663:113-121. [PMID: 37121121 DOI: 10.1016/j.bbrc.2023.03.085] [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: 02/07/2023] [Revised: 03/15/2023] [Accepted: 03/31/2023] [Indexed: 05/02/2023]
Abstract
Retinal ischemic disease is a major type of retinal diseases causing vision loss. Identifying the molecular mechanisms mediating the retinal ischemia-reperfusion (RIR) is the key to targeted intervention. In this study, we performed RNA-seq analysis of the retinal tissues of a retinal ischemia-reperfusion model of Sprague-Dawley (SD) rats, followed by differential gene expression analysis, gene ontology (GO) enrichment analysis, and protein-protein interaction (PPI) analysis. After studying we found that: The major biological processes affected after RIR was the regulation of vascular development. PPI analysis unveiled a regulatory module in which Platelet Derived Growth Factor Receptor Beta (PDGFRB) was upregulated. In the RIR cell model of human retinal microvascular endothelial cells (HRCEC) induced by oxygen-glucose deprivation/reperfusion (OGD/R), silencing PDGFRB at least partially rescued the detrimental effect on cell proliferation and in vitro angiogenic ability. In the rat model of RIR, the administration of PDGFR inhibitor alleviated the damages in the retinal microvascular system. Besides, we further demonstrated the protective effect of procyanidin against RIR induced damages in both the cell and animal model by dampening the overexpression of PDGFRB. Together, our data indicate that the upregulation of PDGFRB contributes to RIR-induced damages in retinal microvascular system, which provides a targetable strategy for therapeutic intervention.
Collapse
Affiliation(s)
- Juanjuan Li
- Affiliated Hospital of Yunnan University (Second People's Hospital of Yunnan Province, Yunnan Eye Hospital), No.176, Qing nian Road, Kunming city, Yunnan Province, 650021, China
| | - Chen Chen
- Affiliated Hospital of Yunnan University (Second People's Hospital of Yunnan Province, Yunnan Eye Hospital), No.176, Qing nian Road, Kunming city, Yunnan Province, 650021, China
| | - Liwei Zhang
- Affiliated Hospital of Yunnan University (Second People's Hospital of Yunnan Province, Yunnan Eye Hospital), No.176, Qing nian Road, Kunming city, Yunnan Province, 650021, China
| | - Yuling Ren
- Affiliated Hospital of Yunnan University (Second People's Hospital of Yunnan Province, Yunnan Eye Hospital), No.176, Qing nian Road, Kunming city, Yunnan Province, 650021, China
| | - Hua Li
- Affiliated Hospital of Yunnan University (Second People's Hospital of Yunnan Province, Yunnan Eye Hospital), No.176, Qing nian Road, Kunming city, Yunnan Province, 650021, China.
| |
Collapse
|
38
|
Evaluation of Retinal Vascularity Index in Patients with COVID-19: A Case-Control Study. Ophthalmol Ther 2023; 12:879-894. [PMID: 36547863 PMCID: PMC9774073 DOI: 10.1007/s40123-022-00630-8] [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: 08/14/2022] [Accepted: 11/30/2022] [Indexed: 12/24/2022] Open
Abstract
INTRODUCTION The purpose of this study was to evaluate the impact of COVID-19 infection on retinal microvasculature by topographically mapping the retinal arteriole-to-venule ratio (AVR). METHODS In a comparative cross-sectional case-control study, fundus photos were obtained in COVID-19-infected patients and healthy controls. AVT was measured over 16 points across the retina using retinal vascularity index (RVI)-a novel semi-automated computerized parameter based on retinal vasculature. RESULTS A total of 51 COVID-19-positive patients and 65 healthy controls were enrolled in the study. Overall, the mean RVI of all 16 points across the retina was 0.34 ± 0.02 in patients with COVID-19 and 0.33 ± 0.02 in control subjects (p = 0.64). Out of the 16 points being measured, three points had a statistically significant greater value in patients with COVID compared to normal controls. CONCLUSION Localised greater RVI values were found in some of the points in COVID-19-positive patients, which likely indicates a more focal change of the vasculature.
Collapse
|
39
|
Burganova G, Schonblum A, Sakhneny L, Epshtein A, Wald T, Tzaig M, Landsman L. Pericytes modulate islet immune cells and insulin secretion through Interleukin-33 production in mice. Front Endocrinol (Lausanne) 2023; 14:1142988. [PMID: 36967785 PMCID: PMC10034381 DOI: 10.3389/fendo.2023.1142988] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Accepted: 02/21/2023] [Indexed: 03/12/2023] Open
Abstract
Introduction Immune cells were recently shown to support β-cells and insulin secretion. However, little is known about how islet immune cells are regulated to maintain glucose homeostasis. Administration of various cytokines, including Interleukin-33 (IL-33), was shown to influence β-cell function. However, the role of endogenous, locally produced IL-33 in pancreatic function remains unknown. Here, we show that IL-33, produced by pancreatic pericytes, is required for glucose homeostasis. Methods To characterize pancreatic IL-33 production, we employed gene expression, flow cytometry, and immunofluorescence analyses. To define the role of this cytokine, we employed transgenic mouse systems to delete the Il33 gene selectively in pancreatic pericytes, in combination with the administration of recombinant IL-33. Glucose response was measured in vivo and in vitro, and morphometric and molecular analyses were used to measure β-cell mass and gene expression. Immune cells were analyzed by flow cytometry. Resuts Our results show that pericytes are the primary source of IL-33 in the pancreas. Mice lacking pericytic IL-33 were glucose intolerant due to impaired insulin secretion. Selective loss of pericytic IL-33 was further associated with reduced T and dendritic cell numbers in the islets and lower retinoic acid production by islet macrophages. Discussion Our study demonstrates the importance of local, pericytic IL-33 production for glucose regulation. Additionally, it proposes that pericytes regulate islet immune cells to support β-cell function in an IL-33-dependent manner. Our study reveals an intricate cellular network within the islet niche.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Limor Landsman
- Department of Cell and Developmental Biology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| |
Collapse
|
40
|
Kamiya E, Morita A, Mori A, Sakamoto K, Nakahara T. The process of methylglyoxal-induced retinal capillary endothelial cell degeneration in rats. Microvasc Res 2023; 146:104455. [PMID: 36396077 DOI: 10.1016/j.mvr.2022.104455] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Revised: 10/28/2022] [Accepted: 11/05/2022] [Indexed: 11/16/2022]
Abstract
Methylglyoxal, a highly reactive dicarbonyl compound, is increased and accumulated in patients with diabetic mellitus. Methylglyoxal forms advanced glycation end products (AGE), contributing to the pathogenesis of diabetic complications, including diabetic retinopathy. Recent studies have shown that methylglyoxal induces diabetic retinopathy-like abnormalities in retinal vasculature. In this study, we investigated the processes and mechanisms of methylglyoxal-induced retinal capillary endothelial cell degeneration in rats. Morphological changes in vascular components (endothelial cells, pericytes, and basement membranes) were assessed in the retinas 2, 7, and 14 days after intravitreal injection of methylglyoxal. Intravitreal methylglyoxal injection induced retinal capillary endothelial cell degeneration in a dose- and time-dependent manner. Changes in the shape and distribution of pericytes occurred before the initiation of capillary regression in the retinas of methylglyoxal-injected eyes. The receptor for AGEs (RAGEs) antagonist FPS-ZM1, and the matrix metalloproteinase (MMP) inhibitor GM6001 significantly attenuated methylglyoxal-induced capillary endothelial cell degeneration. FPS-ZM1 failed to prevent pathological changes in pericytes in methylglyoxal-injected eyes. In situ zymography revealed that MMP activity was enhanced at sites of blood vessels with reduced pericyte coverage in methylglyoxal-injected eyes. These results suggest that intravitreal methylglyoxal injection induces pathological changes in pericytes before the initiation of capillary endothelial cell degeneration via an AGE-RAGE-independent pathway. The capillary endothelial cell degeneration is mediated by activating the AGE-RAGE pathway and increasing MMP activity in endothelial cells by impairing pericyte function in the retina.
Collapse
Affiliation(s)
- Erika Kamiya
- Department of Molecular Pharmacology, Kitasato University School of Pharmaceutical Sciences, 5-9-1 Shirokane, Minato-ku, Tokyo 108-8641, Japan
| | - Akane Morita
- Department of Molecular Pharmacology, Kitasato University School of Pharmaceutical Sciences, 5-9-1 Shirokane, Minato-ku, Tokyo 108-8641, Japan
| | - Asami Mori
- Department of Molecular Pharmacology, Kitasato University School of Pharmaceutical Sciences, 5-9-1 Shirokane, Minato-ku, Tokyo 108-8641, Japan
| | - Kenji Sakamoto
- Department of Molecular Pharmacology, Kitasato University School of Pharmaceutical Sciences, 5-9-1 Shirokane, Minato-ku, Tokyo 108-8641, Japan
| | - Tsutomu Nakahara
- Department of Molecular Pharmacology, Kitasato University School of Pharmaceutical Sciences, 5-9-1 Shirokane, Minato-ku, Tokyo 108-8641, Japan.
| |
Collapse
|
41
|
Shen LL, Mangalesh S, Michalak SM, McGeehan B, Sarin N, Finkle J, Winter KP, Tran-Viet D, Benner EJ, Vajzovic L, Freedman SF, Younge N, Cotten CM, El-Dairi M, Ying GS, Toth C. Associations between systemic health and retinal nerve fibre layer thickness in preterm infants at 36 weeks postmenstrual age. Br J Ophthalmol 2023; 107:242-247. [PMID: 34389548 PMCID: PMC8858642 DOI: 10.1136/bjophthalmol-2021-319254] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Accepted: 07/30/2021] [Indexed: 01/25/2023]
Abstract
BACKGROUND/AIMS Neonatal insults from systemic diseases have been implicated in the pathway of impaired neurodevelopment in preterm infants. We aimed to investigate the associations between systemic health factors and retinal nerve fibre layer (RNFL) thickness in preterm infants. METHODS We prospectively enrolled infants and imaged both eyes at 36±1 weeks postmenstrual age (PMA) using a hand-held optical coherence tomography system at the bedside in the Duke intensive care nurseries. We evaluated associations between RNFL thickness and 29 systemic health factors using univariable and multivariable regression models. RESULTS 83 infants with RNFL thickness measures were included in this study. Based on the multivariable model, RNFL thickness was positively associated with infant weight at imaging and was negatively associated with sepsis/necrotising enterocolitis (NEC). RNFL thickness was 10.4 µm (95% CI -15.9 to -4.9) lower in infants with than without sepsis/NEC in the univariable analysis (p<0.001). This difference remained statistically significant after adjustment for confounding variables in various combinations (birth weight, birthweight percentile, gestational age, infant weight at imaging and growth velocity). A 250 g increase in infant weight at imaging was associated with a 3.1 µm (95% CI 2.1 to 4.2) increase in RNFL thickness in the univariable analysis (p<0.001). CONCLUSIONS Low infant weight and sepsis/NEC were independently associated with thinner RNFL in preterm infants at 36 weeks PMA. To our knowledge, this study is the first to suggest that sepsis/NEC may affect retinal neurodevelopment. Future longitudinal studies are needed to investigate this relationship further.
Collapse
Affiliation(s)
- Liangbo L Shen
- Department of Ophthalmology, University of California San Francisco, San Francisco, California, USA
| | - Shwetha Mangalesh
- Department of Ophthalmology, Duke University School of Medicine, Durham, North Carolina, USA
| | - Suzanne M Michalak
- Department of Ophthalmology, Duke University School of Medicine, Durham, North Carolina, USA
| | - Brendan McGeehan
- Department of Ophthalmology, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Neeru Sarin
- Department of Ophthalmology, Duke University School of Medicine, Durham, North Carolina, USA
| | - Joanne Finkle
- Department of Pediatrics, Duke University School of Medicine, Durham, North Carolina, USA
| | - Katrina P Winter
- Department of Ophthalmology, Duke University School of Medicine, Durham, North Carolina, USA
| | - Du Tran-Viet
- Department of Ophthalmology, Duke University School of Medicine, Durham, North Carolina, USA
| | - Eric J Benner
- Department of Pediatrics, Duke University School of Medicine, Durham, North Carolina, USA
| | - Lejla Vajzovic
- Department of Ophthalmology, Duke University School of Medicine, Durham, North Carolina, USA
| | - Sharon F Freedman
- Department of Ophthalmology, Duke University School of Medicine, Durham, North Carolina, USA
- Department of Pediatrics, Duke University School of Medicine, Durham, North Carolina, USA
| | - Noelle Younge
- Department of Pediatrics, Duke University School of Medicine, Durham, North Carolina, USA
| | - C Michael Cotten
- Department of Pediatrics, Duke University School of Medicine, Durham, North Carolina, USA
| | - Mays El-Dairi
- Department of Ophthalmology, Duke University School of Medicine, Durham, North Carolina, USA
| | - Gui-Shuang Ying
- Department of Ophthalmology, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Cynthia Toth
- Department of Ophthalmology, Duke University School of Medicine, Durham, North Carolina, USA
- Department of Biomedical Engineering, Duke University, Durham, North Carolina, USA
| |
Collapse
|
42
|
Different Contacted Cell Types Contribute to Acquiring Different Properties in Brain Microglial Cells upon Intercellular Interaction. Int J Mol Sci 2023; 24:ijms24021774. [PMID: 36675286 PMCID: PMC9861207 DOI: 10.3390/ijms24021774] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Revised: 01/12/2023] [Accepted: 01/12/2023] [Indexed: 01/18/2023] Open
Abstract
Microglial cells (MGs), originally derived from progenitor cells in a yolk sac during early development, are glial cells located in a physiological and pathological brain. Since the brain contains various cell types, MGs could frequently interact with different cells, such as astrocytes (ACs), pericytes (PCs), and endothelial cells (ECs). However, how microglial traits are regulated via cell-cell interactions by ACs, PCs, or ECs and how they are different depending on the contacted cell types is unclear. This study aimed to clarify these questions by coculturing MGs with ACs, PCs, or ECs using mouse brain-derived cells, and microglial phenotypic changes were investigated under culture conditions that enabled direct cell-cell contact. Our results showed that ACs or PCs dose-dependently increased the number of MG, while ECs decreased it. Microarray and gene ontology analysis showed that cell fate-related genes (e.g., cell cycle, proliferation, growth, death, and apoptosis) of MGs were altered after a cell-cell contact with ACs, PCs, and ECs. Notably, microarray analysis showed that several genes, such as gap junction protein alpha 1 (Gja1), were prominently upregulated in MGs after coincubation with ACs, PCs, or ECs, regardless of cell types. Similarly, immunohistochemistry showed that an increased Gja1 expression was observed in MGs after coincubation with ACs, PCs, or ECs. Immunofluorescent and fluorescence-activated cell sorting analysis also showed that calcein-AM was transferred into MGs after coincubation with ACs, PCs, or ECs, confirming that intercellular interactions occurred between these cells. However, while Gja1 inhibition reduced the number of MGs after coincubation with ACs and PCs, this was increased after coincubation with ECs; this indicates that ACs and PCs positively regulate microglial numbers via Gja1, while ECs decrease it. Results show that ACs, PCs, or ECs exert both common and specific cell type-dependent effects on MGs through intercellular interactions. These findings also suggest that brain microglial phenotypes are different depending on their surrounding cell types, such as ACs, PCs, or ECs.
Collapse
|
43
|
Otsuka T, Masuda T, Takahashi Y, Suzuki A, Uemura A, Arakawa R, Okabe T, Naito A. Effect of triamcinolone acetonide on retinal inflammation and angiogenesis induced by pericyte depletion in mouse. J Pharmacol Sci 2023; 151:28-36. [DOI: 10.1016/j.jphs.2022.10.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2022] [Revised: 10/10/2022] [Accepted: 10/21/2022] [Indexed: 11/18/2022] Open
|
44
|
Hattori Y. The Multiple Roles of Pericytes in Vascular Formation and Microglial Functions in the Brain. Life (Basel) 2022; 12:1835. [PMID: 36362989 PMCID: PMC9699346 DOI: 10.3390/life12111835] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Revised: 11/07/2022] [Accepted: 11/08/2022] [Indexed: 10/15/2023] Open
Abstract
In the capillary walls, vascular endothelial cells are covered with mural cells, such as smooth muscle cells and pericytes. Although pericytes had been thought to play simply a structural role, emerging evidence has highlighted their multiple functions in the embryonic, postnatal, and adult brain. As the central nervous system (CNS) develops, the brain's vascular structure gradually matures into a hierarchical network, which is crucial for the proper development of neural lineage cells by providing oxygen and nutrients. Pericytes play an essential role in vascular formation and regulate blood‒brain barrier (BBB) integrity as a component of the neurovascular unit (NVU), in collaboration with other cells, such as vascular endothelial cells, astrocytes, neurons, and microglia. Microglia, the resident immune cells of the CNS, colonize the brain at embryonic day (E) 9.5 in mice. These cells not only support the development and maturation of neural lineage cells but also help in vascular formation through their extensive migration. Recent studies have demonstrated that pericytes directly contact microglia in the CNS, and their interactions have a profound effect on physiological and pathological aspects. This review summarizes the function of pericytes, focusing on the interplay between pericytes and microglia.
Collapse
Affiliation(s)
- Yuki Hattori
- Department of Anatomy and Cell Biology, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan
| |
Collapse
|
45
|
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: 29] [Impact Index Per Article: 9.7] [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
|
46
|
Ren J, Zhang S, Pan Y, Jin M, Li J, Luo Y, Sun X, Li G. Diabetic retinopathy: Involved cells, biomarkers, and treatments. Front Pharmacol 2022; 13:953691. [PMID: 36016568 PMCID: PMC9396039 DOI: 10.3389/fphar.2022.953691] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Accepted: 07/06/2022] [Indexed: 11/13/2022] Open
Abstract
Diabetic retinopathy (DR), a leading cause of vision loss and blindness worldwide, is caused by retinal neurovascular unit dysfunction, and its cellular pathology involves at least nine kinds of retinal cells, including photoreceptors, horizontal and bipolar cells, amacrine cells, retinal ganglion cells, glial cells (Müller cells, astrocytes, and microglia), endothelial cells, pericytes, and retinal pigment epithelial cells. Its mechanism is complicated and involves loss of cells, inflammatory factor production, neovascularization, and BRB impairment. However, the mechanism has not been completely elucidated. Drug treatment for DR has been gradually advancing recently. Research on potential drug targets relies upon clear information on pathogenesis and effective biomarkers. Therefore, we reviewed the recent literature on the cellular pathology and the diagnostic and prognostic biomarkers of DR in terms of blood, protein, and clinical and preclinical drug therapy (including synthesized molecules and natural molecules). This review may provide a theoretical basis for further DR research.
Collapse
Affiliation(s)
- Jiahui Ren
- Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
- Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Beijing, China
- Key Laboratory of Bioactive Substances and Resource Utilization of Chinese Herbal Medicine, Ministry of Education, Beijing, China
- Yunnan Branch, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences and Peking Union Medical College, Jinghong, China
- Yunnan Key Laboratory of Southern Medicine Utilization, Kunming, China
| | - Shuxia Zhang
- Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
- Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Beijing, China
- Key Laboratory of Bioactive Substances and Resource Utilization of Chinese Herbal Medicine, Ministry of Education, Beijing, China
| | - Yunfeng Pan
- Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
- Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Beijing, China
- Key Laboratory of Bioactive Substances and Resource Utilization of Chinese Herbal Medicine, Ministry of Education, Beijing, China
| | - Meiqi Jin
- Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
- Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Beijing, China
- Key Laboratory of Bioactive Substances and Resource Utilization of Chinese Herbal Medicine, Ministry of Education, Beijing, China
| | - Jiaxin Li
- Yunnan Key Laboratory of Southern Medicine Utilization, Kunming, China
- College of Pharmacy, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Yun Luo
- Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
- Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Beijing, China
- Key Laboratory of Bioactive Substances and Resource Utilization of Chinese Herbal Medicine, Ministry of Education, Beijing, China
- *Correspondence: Yun Luo, ; Xiaobo Sun , ; Guang Li,
| | - Xiaobo Sun
- Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
- Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Beijing, China
- Key Laboratory of Bioactive Substances and Resource Utilization of Chinese Herbal Medicine, Ministry of Education, Beijing, China
- *Correspondence: Yun Luo, ; Xiaobo Sun , ; Guang Li,
| | - Guang Li
- Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
- Yunnan Branch, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences and Peking Union Medical College, Jinghong, China
- Yunnan Key Laboratory of Southern Medicine Utilization, Kunming, China
- *Correspondence: Yun Luo, ; Xiaobo Sun , ; Guang Li,
| |
Collapse
|
47
|
Orlich MM, Diéguez-Hurtado R, Muehlfriedel R, Sothilingam V, Wolburg H, Oender CE, Woelffing P, Betsholtz C, Gaengel K, Seeliger M, Adams RH, Nordheim A. Mural Cell SRF Controls Pericyte Migration, Vessel Patterning and Blood Flow. Circ Res 2022; 131:308-327. [PMID: 35862101 PMCID: PMC9348820 DOI: 10.1161/circresaha.122.321109] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Pericytes and vascular smooth muscle cells, collectively known as mural cells, are recruited through PDGFB (platelet-derived growth factor B)-PDGFRB (platelet-derived growth factor receptor beta) signaling. MCs are essential for vascular integrity, and their loss has been associated with numerous diseases. Most of this knowledge is based on studies in which MCs are insufficiently recruited or fully absent upon inducible ablation. In contrast, little is known about the physiological consequences that result from impairment of specific MC functions. Here, we characterize the role of the transcription factor SRF (serum response factor) in MCs and study its function in developmental and pathological contexts.
Collapse
Affiliation(s)
- Michael M. Orlich
- Department of Molecular Biology, Interfaculty Institute for Cell Biology, University of Tuebingen, Germany (M.M.O., C.E.O., P.W., A.N.)
- International Max Planck Research School (IMPRS) “From Molecules to Organisms,” Tuebingen, Germany (M.M.O., A.N.)
- Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala University, Sweden (M.M.O., C.B., K.G.)
- Now with Rudbeck Laboratory C11, Dag Hammarskjölds Väg 20, 751 85 Uppsala, Sweden (M.M.O.)
| | - Rodrigo Diéguez-Hurtado
- Department of Tissue Morphogenesis, Max Planck Institute for Molecular Biomedicine, Muenster, Germany (R.D.-H., R.H.A.)
- Faculty of Medicine, University of Muenster, Muenster, Germany (R.D.-H., R.H.A.)
| | - Regine Muehlfriedel
- Institute for Ophthalmic Research, Centre for Ophthalmology, University Clinic Tuebingen (UKT), Germany. (R.M., V.S., M.S.)
| | - Vithiyanjali Sothilingam
- Institute for Ophthalmic Research, Centre for Ophthalmology, University Clinic Tuebingen (UKT), Germany. (R.M., V.S., M.S.)
| | - Hartwig Wolburg
- Department of General Pathology and Pathological Anatomy, Institute of Pathology and Neuropathology, University Clinic Tuebingen (UKT), Germany. (H.W.)
| | - Cansu Ebru Oender
- Department of Molecular Biology, Interfaculty Institute for Cell Biology, University of Tuebingen, Germany (M.M.O., C.E.O., P.W., A.N.)
| | - Pascal Woelffing
- Department of Molecular Biology, Interfaculty Institute for Cell Biology, University of Tuebingen, Germany (M.M.O., C.E.O., P.W., A.N.)
| | - Christer Betsholtz
- Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala University, Sweden (M.M.O., C.B., K.G.)
| | - Konstantin Gaengel
- Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala University, Sweden (M.M.O., C.B., K.G.)
| | - Mathias Seeliger
- Institute for Ophthalmic Research, Centre for Ophthalmology, University Clinic Tuebingen (UKT), Germany. (R.M., V.S., M.S.)
| | - Ralf H. Adams
- Department of Tissue Morphogenesis, Max Planck Institute for Molecular Biomedicine, Muenster, Germany (R.D.-H., R.H.A.)
- Faculty of Medicine, University of Muenster, Muenster, Germany (R.D.-H., R.H.A.)
| | - Alfred Nordheim
- Department of Molecular Biology, Interfaculty Institute for Cell Biology, University of Tuebingen, Germany (M.M.O., C.E.O., P.W., A.N.)
- International Max Planck Research School (IMPRS) “From Molecules to Organisms,” Tuebingen, Germany (M.M.O., A.N.)
| |
Collapse
|
48
|
Galoppin M, Kari S, Soldati S, Pal A, Rival M, Engelhardt B, Astier A, Thouvenot E. Full spectrum of vitamin D immunomodulation in multiple sclerosis: mechanisms and therapeutic implications. Brain Commun 2022; 4:fcac171. [PMID: 35813882 PMCID: PMC9260308 DOI: 10.1093/braincomms/fcac171] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Revised: 05/03/2022] [Accepted: 06/28/2022] [Indexed: 11/17/2022] Open
Abstract
Vitamin D deficiency has been associated with the risk of multiple sclerosis, disease activity and progression. Results from in vitro experiments, animal models and analysis of human samples from randomized controlled trials provide comprehensive data illustrating the pleiotropic actions of Vitamin D on the immune system. They globally result in immunomodulation by decreasing differentiation of effector T and B cells while promoting regulatory subsets. Vitamin D also modulates innate immune cells such as macrophages, monocytes and dendritic cells, and acts at the level of the blood–brain barrier reducing immune cell trafficking. Vitamin D exerts additional activity within the central nervous system reducing microglial and astrocytic activation. The immunomodulatory role of Vitamin D detected in animal models of multiple sclerosis has suggested its potential therapeutic use for treating multiple sclerosis. In this review, we focus on recent published data describing the biological effects of Vitamin D in animal models of multiple sclerosis on immune cells, blood–brain barrier function, activation of glial cells and its potential neuroprotective effects. Based on the current knowledge, we also discuss optimization of therapeutic interventions with Vitamin D in patients with multiple sclerosis, as well as new technologies allowing in-depth analysis of immune cell regulations by vitamin D.
Collapse
Affiliation(s)
- Manon Galoppin
- IGF, University Montpellier, CNRS, INSERM , Montpellier , France
| | - Saniya Kari
- Toulouse Institute for Infectious and Inflammatory Diseases (Infinity), INSERM UMR1291 – CNRS UMR5051 – Université Toulouse III , 31024 Toulouse cedex 3 , France
| | - Sasha Soldati
- Theodor Kocher Institute, University of Bern , Bern , Switzerland
| | - Arindam Pal
- Theodor Kocher Institute, University of Bern , Bern , Switzerland
| | - Manon Rival
- IGF, University Montpellier, CNRS, INSERM , Montpellier , France
- Department of Neurology, Nîmes University Hospital, University Montpellier , Nîmes , France
| | | | - Anne Astier
- Toulouse Institute for Infectious and Inflammatory Diseases (Infinity), INSERM UMR1291 – CNRS UMR5051 – Université Toulouse III , 31024 Toulouse cedex 3 , France
| | - Eric Thouvenot
- IGF, University Montpellier, CNRS, INSERM , Montpellier , France
- Department of Neurology, Nîmes University Hospital, University Montpellier , Nîmes , France
| |
Collapse
|
49
|
Barkaway A, Attwell D, Korte N. Immune-vascular mural cell interactions: consequences for immune cell trafficking, cerebral blood flow, and the blood-brain barrier. NEUROPHOTONICS 2022; 9:031914. [PMID: 35581998 PMCID: PMC9107322 DOI: 10.1117/1.nph.9.3.031914] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Accepted: 04/13/2022] [Indexed: 06/15/2023]
Abstract
Brain barriers are crucial sites for cerebral energy supply, waste removal, immune cell migration, and solute exchange, all of which maintain an appropriate environment for neuronal activity. At the capillary level, where the largest area of brain-vascular interface occurs, pericytes adjust cerebral blood flow (CBF) by regulating capillary diameter and maintain the blood-brain barrier (BBB) by suppressing endothelial cell (EC) transcytosis and inducing tight junction expression between ECs. Pericytes also limit the infiltration of circulating leukocytes into the brain where resident microglia confine brain injury and provide the first line of defence against invading pathogens. Brain "waste" is cleared across the BBB into the blood, phagocytosed by microglia and astrocytes, or removed by the flow of cerebrospinal fluid (CSF) through perivascular routes-a process driven by respiratory motion and the pulsation of the heart, arteriolar smooth muscle, and possibly pericytes. "Dirty" CSF exits the brain and is probably drained around olfactory nerve rootlets and via the dural meningeal lymphatic vessels and possibly the skull bone marrow. The brain is widely regarded as an immune-privileged organ because it is accessible to few antigen-primed leukocytes. Leukocytes enter the brain via the meninges, the BBB, and the blood-CSF barrier. Advances in genetic and imaging tools have revealed that neurological diseases significantly alter immune-brain barrier interactions in at least three ways: (1) the brain's immune-privileged status is compromised when pericytes are lost or lymphatic vessels are dysregulated; (2) immune cells release vasoactive molecules to regulate CBF, modulate arteriole stiffness, and can plug and eliminate capillaries which impairs CBF and possibly waste clearance; and (3) immune-vascular interactions can make the BBB leaky via multiple mechanisms, thus aggravating the influx of undesirable substances and cells. Here, we review developments in these three areas and briefly discuss potential therapeutic avenues for restoring brain barrier functions.
Collapse
Affiliation(s)
- Anna Barkaway
- University College London, Department of Neuroscience, Physiology and Pharmacology, London, United Kingdom
| | - David Attwell
- University College London, Department of Neuroscience, Physiology and Pharmacology, London, United Kingdom
| | - Nils Korte
- University College London, Department of Neuroscience, Physiology and Pharmacology, London, United Kingdom
| |
Collapse
|
50
|
Cheung CMG, Fawzi A, Teo KY, Fukuyama H, Sen S, Tsai WS, Sivaprasad S. Diabetic macular ischaemia- a new therapeutic target? Prog Retin Eye Res 2022; 89:101033. [PMID: 34902545 PMCID: PMC11268431 DOI: 10.1016/j.preteyeres.2021.101033] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2021] [Revised: 11/25/2021] [Accepted: 12/01/2021] [Indexed: 12/21/2022]
Abstract
Diabetic macular ischaemia (DMI) is traditionally defined and graded based on the angiographic evidence of an enlarged and irregular foveal avascular zone. However, these anatomical changes are not surrogate markers for visual impairment. We postulate that there are vascular phenotypes of DMI based on the relative perfusion deficits of various retinal capillary plexuses and choriocapillaris. This review highlights several mechanistic pathways, including the role of hypoxia and the complex relation between neurons, glia, and microvasculature. The current animal models are reviewed, with shortcomings noted. Therefore, utilising the advancing technology of optical coherence tomography angiography (OCTA) to identify the reversible DMI phenotypes may be the key to successful therapeutic interventions for DMI. However, there is a need to standardise the nomenclature of OCTA perfusion status. Visual acuity is not an ideal endpoint for DMI clinical trials. New trial endpoints that represent disease progression need to be developed before irreversible vision loss in patients with DMI. Natural history studies are required to determine the course of each vascular and neuronal parameter to define the DMI phenotypes. These DMI phenotypes may also partly explain the development and recurrence of diabetic macular oedema. It is also currently unclear where and how DMI fits into the diabetic retinopathy severity scales, further highlighting the need to better define the progression of diabetic retinopathy and DMI based on both multimodal imaging and visual function. Finally, we discuss a complete set of proposed therapeutic pathways for DMI, including cell-based therapies that may provide restorative potential.
Collapse
Affiliation(s)
- Chui Ming Gemmy Cheung
- Singapore Eye Research Institution, Singapore National Eye Centre, Singapore; Duke-NUS Medical School, National University of Singapore, Singapore
| | | | - Kelvin Yc Teo
- Singapore Eye Research Institution, Singapore National Eye Centre, Singapore
| | | | | | - Wei-Shan Tsai
- NIHR Moorfields Biomedical Research Centre, Moorfields Eye Hospital, London, United Kingdom
| | - Sobha Sivaprasad
- NIHR Moorfields Biomedical Research Centre, Moorfields Eye Hospital, London, United Kingdom.
| |
Collapse
|