1
|
Hayes MN, Cohen-Gogo S, Kee L, Xiong X, Weiss A, Layeghifard M, Ladumor Y, Valencia-Sama I, Rajaselvam A, Kaplan DR, Villani A, Shlien A, Morgenstern DA, Irwin MS. DNA damage response deficiency enhances neuroblastoma progression and sensitivity to combination PARP and ATR inhibition. Cell Rep 2025; 44:115537. [PMID: 40220294 DOI: 10.1016/j.celrep.2025.115537] [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: 10/11/2024] [Revised: 02/03/2025] [Accepted: 03/17/2025] [Indexed: 04/14/2025] Open
Abstract
Sequencing of neuroblastoma (NB) tumors has revealed genetic alterations in genes involved in DNA damage response (DDR) pathways. However, roles for specific alterations of DDR genes in pediatric solid tumors remain poorly understood. To address this, mutations in the DDR pathway including Brca2, Atm, and Palb2 were incorporated into an established zebrafish MYCN transgenic model (Tg(dbh:EGFP-MYCN)). These mutations enhance NB formation and metastasis and result in upregulation of cell-cycle checkpoint and DNA damage repair signatures, revealing molecular vulnerabilities in DDR-deficient NB. DDR gene knockdown in zebrafish and human NB cells increases sensitivity to the poly(ADP-ribose) polymerase (PARP) inhibitor olaparib, and this effect is enhanced by inhibition of the ataxia telangiectasia and rad3-related (ATR) kinase. This work provides in vivo evidence demonstrating that alterations in certain DDR-pathway genes promote aggressive NB and supports combination PARP + ATR inhibitor therapy for NB patients with tumors harboring specific genetic alterations in DDR.
Collapse
Affiliation(s)
- Madeline N Hayes
- Developmental, Stem Cell and Cancer Biology Program, The Hospital for Sick Children, Toronto, ON, Canada; Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada.
| | - Sarah Cohen-Gogo
- Division of Hematology/Oncology, The Hospital for Sick Children, Toronto, ON, Canada; Genetics and Genome Biology Program, The Hospital for Sick Children, Toronto, ON, Canada
| | - Lynn Kee
- Cell Biology Program, The Hospital for Sick Children, Toronto, ON, Canada
| | - Xueting Xiong
- Developmental, Stem Cell and Cancer Biology Program, The Hospital for Sick Children, Toronto, ON, Canada; Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
| | - Alex Weiss
- Developmental, Stem Cell and Cancer Biology Program, The Hospital for Sick Children, Toronto, ON, Canada
| | - Mehdi Layeghifard
- Genetics and Genome Biology Program, The Hospital for Sick Children, Toronto, ON, Canada
| | - Yagnesh Ladumor
- Cell Biology Program, The Hospital for Sick Children, Toronto, ON, Canada; Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada
| | | | - Anisha Rajaselvam
- Cell Biology Program, The Hospital for Sick Children, Toronto, ON, Canada; Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada
| | - David R Kaplan
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada; Neurosciences and Mental Health Program, The Hospital for Sick Children, Toronto, ON, Canada
| | - Anita Villani
- Division of Hematology/Oncology, The Hospital for Sick Children, Toronto, ON, Canada; Department of Pediatrics, University of Toronto, Toronto, ON, Canada
| | - Adam Shlien
- Genetics and Genome Biology Program, The Hospital for Sick Children, Toronto, ON, Canada; Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
| | - Daniel A Morgenstern
- Division of Hematology/Oncology, The Hospital for Sick Children, Toronto, ON, Canada; Department of Pediatrics, University of Toronto, Toronto, ON, Canada
| | - Meredith S Irwin
- Cell Biology Program, The Hospital for Sick Children, Toronto, ON, Canada; Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada; Department of Pediatrics, University of Toronto, Toronto, ON, Canada; Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada; Department of Pediatrics, The Hospital for Sick Children, Toronto, ON, Canada.
| |
Collapse
|
2
|
Tang Y, Cheng C, Ding R, Qian J, Liu M, Guo Y, Li Q. MSC exosomes and MSC exosomes loaded with LncRNA H19 as nanotherapeutics regulate the neurogenetic potential of Müller Glial Cells in dry age-related macular degeneration. Free Radic Biol Med 2025; 231:178-192. [PMID: 40015462 DOI: 10.1016/j.freeradbiomed.2025.02.039] [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: 01/17/2025] [Revised: 02/13/2025] [Accepted: 02/24/2025] [Indexed: 03/01/2025]
Abstract
In retinal degeneration diseases such as dry age-related macular degeneration (AMD), Müller Glial Cells (MGCs) in mammals undergo a process of reactive gliosis leading to the progression of dry AMD. Here, It is demonstrated that exosomes derived from mesenchymal stem cells (MSC exosomes) and MSC exosomes loaded with LncRNA H19, acting as nanotherapeutics, can be regulated by MGCs in dry AMD. In the in vivo study, MSC exosomes were administered via intravitreal injection. MSC exosomes effectively redirected MGCs from gliosis to dedifferentiation and alleviated MGCs-to-epithelial transition by inhibiting oxidative stress in mice with dry AMD induced by NaIO3. In the in vitro study, MSC exosomes promoted MGCs dedifferentiation by activating Wnt/β-catenin signaling pathway and prevented oxidative stress-induced MGCs gliosis and MGCs-to-epithelial transition by inhibiting TGFβ1 signaling pathway. MSC exosomes loaded with LncRNA H19 enhanced the activation of Wnt/β-catenin signaling pathway and the inhibition of the TGFβ1 signaling pathway compared with MSC exosomes. These results suggest that MSC exosomes regulate the neurogenetic potential of MGCs by redirecting MGCs from gliosis to dedifferentiation and alleviating the transformation of MGCs to epithelial cells through regulating oxidative stress. Regulating LncRNA H19 in MGCs to promote mammalian retinal regeneration in dry AMD was suggested for the first time.
Collapse
Affiliation(s)
- Yue Tang
- China Pharmaceutical University, Nanjing, 211198, PR China.
| | - Caiyi Cheng
- China Pharmaceutical University, Nanjing, 211198, PR China
| | - Rui Ding
- China Pharmaceutical University, Nanjing, 211198, PR China
| | - Jingyuan Qian
- China Pharmaceutical University, Nanjing, 211198, PR China
| | - Min Liu
- China Pharmaceutical University, Nanjing, 211198, PR China
| | - Yuzun Guo
- China Pharmaceutical University, Nanjing, 211198, PR China
| | - Qian Li
- China Pharmaceutical University, Nanjing, 211198, PR China.
| |
Collapse
|
3
|
Wang Y, Tong X, Xiao Y, Wang Y, Hu W, Lu W, Chen Y, Li J, Gao W, Gao H, Tian Y, Dai S, Feng Y. Regulating Integrin β1 to Restore Gonadotropin-Releasing Hormone-Tanycyte Unit Function in Polycystic Ovary Syndrome-Related Hypothalamic Dysregulation. RESEARCH (WASHINGTON, D.C.) 2025; 8:0619. [PMID: 39975575 PMCID: PMC11836200 DOI: 10.34133/research.0619] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/13/2024] [Revised: 01/27/2025] [Accepted: 01/29/2025] [Indexed: 02/21/2025]
Abstract
Excessive gonadotropin-releasing hormone (GnRH) is considered to be an initiating factor in the etiology of polycystic ovary syndrome (PCOS). GnRH neuronal axons terminate at the hypothalamic arcuate nucleus and median eminence, where tanycytes, specialized glial cells, have been proposed to modulate GnRH secretion through plasticity. However, the precise role of the "GnRH-tanycyte unit" during the pathological state of PCOS has not been thoroughly explored. In this study, we demonstrated the architecture and distribution of GnRH neurons and tanycytes. In PCOS-like mice, retracted tanycyte processes and dysregulated GnRH-tanycyte unit may create an environment conducive to the excessive secretion of GnRH and subsequent reproductive endocrine dysfunction. Mechanistically, excessive androgens impair hypothalamic neuroglial homeostasis by acting through the androgen receptor (AR) and its downstream target integrin β1 (Itgb1), thereby suppressing the FAK/TGF-βR1/Smad2 signaling pathway. Both selective deletion of AR and overexpression of Itgb1 in tanycytes counteracted the detrimental effects of androgens, alleviating endocrine dysfunction. Collectively, this study highlights the alterations in the GnRH-tanycyte unit mediated by androgen/AR/Itgb1 signaling and provides a novel perspective for developing therapies for hypothalamic hormone secretion disorders by maintaining solid neuroglial structures in the brain.
Collapse
Affiliation(s)
- Yu Wang
- Department of Integrative Medicine and Neurobiology, School of Basic Medical Sciences, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science,
Fudan University, Shanghai 200032, China
- Shanghai Key Laboratory of Acupuncture Mechanism and Acupoint Function, Shanghai Institute of Acupuncture and Moxibustion, Shanghai 200433, China
| | - Xiaoyu Tong
- Department of Integrative Medicine and Neurobiology, School of Basic Medical Sciences, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science,
Fudan University, Shanghai 200032, China
- Shanghai Key Laboratory of Acupuncture Mechanism and Acupoint Function, Shanghai Institute of Acupuncture and Moxibustion, Shanghai 200433, China
| | - Yan Xiao
- Department of Integrative Medicine and Neurobiology, School of Basic Medical Sciences, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science,
Fudan University, Shanghai 200032, China
- Shanghai Key Laboratory of Acupuncture Mechanism and Acupoint Function, Shanghai Institute of Acupuncture and Moxibustion, Shanghai 200433, China
| | - Yicong Wang
- Department of Integrative Medicine and Neurobiology, School of Basic Medical Sciences, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science,
Fudan University, Shanghai 200032, China
- Shanghai Key Laboratory of Acupuncture Mechanism and Acupoint Function, Shanghai Institute of Acupuncture and Moxibustion, Shanghai 200433, China
| | - Wei Hu
- Department of Integrative Medicine and Neurobiology, School of Basic Medical Sciences, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science,
Fudan University, Shanghai 200032, China
- Shanghai Key Laboratory of Acupuncture Mechanism and Acupoint Function, Shanghai Institute of Acupuncture and Moxibustion, Shanghai 200433, China
| | - Wenhan Lu
- Department of Ophthalmology & Visual Science, Eye & ENT Hospital, Shanghai Medical College,
Fudan University, Shanghai, China
| | - Yuning Chen
- Department of Integrative Medicine and Neurobiology, School of Basic Medical Sciences, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science,
Fudan University, Shanghai 200032, China
- Shanghai Key Laboratory of Acupuncture Mechanism and Acupoint Function, Shanghai Institute of Acupuncture and Moxibustion, Shanghai 200433, China
| | - Jiajia Li
- Department of Integrative Medicine and Neurobiology, School of Basic Medical Sciences, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science,
Fudan University, Shanghai 200032, China
- Shanghai Key Laboratory of Acupuncture Mechanism and Acupoint Function, Shanghai Institute of Acupuncture and Moxibustion, Shanghai 200433, China
| | - Wenhao Gao
- Department of Integrative Medicine and Neurobiology, School of Basic Medical Sciences, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science,
Fudan University, Shanghai 200032, China
- Shanghai Key Laboratory of Acupuncture Mechanism and Acupoint Function, Shanghai Institute of Acupuncture and Moxibustion, Shanghai 200433, China
| | - Hongru Gao
- Department of Integrative Medicine and Neurobiology, School of Basic Medical Sciences, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science,
Fudan University, Shanghai 200032, China
- Shanghai Key Laboratory of Acupuncture Mechanism and Acupoint Function, Shanghai Institute of Acupuncture and Moxibustion, Shanghai 200433, China
| | - Yicheng Tian
- Department of Integrative Medicine and Neurobiology, School of Basic Medical Sciences, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science,
Fudan University, Shanghai 200032, China
- Shanghai Key Laboratory of Acupuncture Mechanism and Acupoint Function, Shanghai Institute of Acupuncture and Moxibustion, Shanghai 200433, China
| | - Sizhe Dai
- Department of Integrative Medicine and Neurobiology, School of Basic Medical Sciences, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science,
Fudan University, Shanghai 200032, China
- Shanghai Key Laboratory of Acupuncture Mechanism and Acupoint Function, Shanghai Institute of Acupuncture and Moxibustion, Shanghai 200433, China
| | - Yi Feng
- Department of Integrative Medicine and Neurobiology, School of Basic Medical Sciences, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science,
Fudan University, Shanghai 200032, China
- Shanghai Key Laboratory of Acupuncture Mechanism and Acupoint Function, Shanghai Institute of Acupuncture and Moxibustion, Shanghai 200433, China
| |
Collapse
|
4
|
Wang Y, Wang Y, Chen Y, Lu W, Tong X, Li J, Gao W, Huang R, Hu W, Feng Y. Distinct mechanisms of electroacupuncture and manual acupuncture in modulating hypothalamic GnRH-tanycyte unit function of polycystic ovary syndrome. Chin Med 2025; 20:18. [PMID: 39910658 DOI: 10.1186/s13020-025-01068-3] [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: 09/04/2024] [Accepted: 01/20/2025] [Indexed: 02/07/2025] Open
Abstract
BACKGROUND Polycystic ovary syndrome (PCOS) is a complex neuroendocrine disorder characterized by dysregulation of the hypothalamus. Both electroacupuncture (EA) and manual acupuncture (MA) have demonstrated therapeutic efficacy in the treatment of PCOS through improvements in hypothalamic function. However, the underlying mechanisms remain poorly understood. Gonadotropin-releasing hormone (GnRH) neurons are pivotal in regulating hypothalamic endocrine function, whereas tanycyte, a specialized glial cell type, potentially contribute to this process. METHODS A dihydrotestosterone (DHT)-induced PCOS-like mouse model was used to investigate the effects of acupuncture. Tissue clearing and three-dimensional (3D) imaging were employed to visualize the hypothalamic GnRH neuronal network and assess postacupuncture modifications. Transcriptome sequencing was performed to identify changes in the gene profiles associated with EA and MA. Rax-CreERT2 transgenic mice were utilized to investigate the molecular targets of EA in tanycytes. RESULTS EA significantly alleviated neuroendocrine dysfunction in PCOS-like mice by restoring the density and coverage of GnRH axonal projections. MA displayed similar therapeutic effects but had less pronounced effects on GnRH axons. Transcriptome analysis revealed distinct mechanisms for these two approaches: EA primarily regulates neuroglial plasticity, whereas MA predominantly targets neurotransmitter regulation. Both EA and MA share a common therapeutic target in the integrin family. Functional studies in Rax-CreERT2 transgenic mice confirmed that Itgb1 plays a critical role in maintaining the balance of hypothalamic GnRH-tanycyte unit during EA treatment. CONCLUSIONS EA exerts therapeutic effects on PCOS by targeting hypothalamic GnRH-tanycyte unit, with Itgb1 identified as a key factor. MA primarily functions through neurotransmitter regulation. These findings highlight potential hypothalamic targets and provide new insights into the distinct mechanisms of EA and MA.
Collapse
Affiliation(s)
- Yu Wang
- Department of Integrative Medicine and Neurobiology, School of Basic Medical Sciences, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science , Fudan University, Shanghai, 200032, China
- Shanghai Key Laboratory of Acupuncture Mechanism and Acupoint Function, Shanghai Institute of Acupuncture and Moxibustion, Fudan University, Shanghai, 200433, China
| | - Yicong Wang
- Department of Integrative Medicine and Neurobiology, School of Basic Medical Sciences, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science , Fudan University, Shanghai, 200032, China
- Shanghai Key Laboratory of Acupuncture Mechanism and Acupoint Function, Shanghai Institute of Acupuncture and Moxibustion, Fudan University, Shanghai, 200433, China
| | - Yuning Chen
- Department of Integrative Medicine and Neurobiology, School of Basic Medical Sciences, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science , Fudan University, Shanghai, 200032, China
- Shanghai Key Laboratory of Acupuncture Mechanism and Acupoint Function, Shanghai Institute of Acupuncture and Moxibustion, Fudan University, Shanghai, 200433, China
| | - Wenhan Lu
- Department of Ophthalmology & Visual Science, Eye & ENT Hospital, Shanghai Medical College, Fudan University, Shanghai, China
| | - Xiaoyu Tong
- Department of Integrative Medicine and Neurobiology, School of Basic Medical Sciences, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science , Fudan University, Shanghai, 200032, China
- Shanghai Key Laboratory of Acupuncture Mechanism and Acupoint Function, Shanghai Institute of Acupuncture and Moxibustion, Fudan University, Shanghai, 200433, China
| | - Jiajia Li
- Department of Integrative Medicine and Neurobiology, School of Basic Medical Sciences, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science , Fudan University, Shanghai, 200032, China
- Shanghai Key Laboratory of Acupuncture Mechanism and Acupoint Function, Shanghai Institute of Acupuncture and Moxibustion, Fudan University, Shanghai, 200433, China
| | - Wenhao Gao
- Department of Integrative Medicine and Neurobiology, School of Basic Medical Sciences, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science , Fudan University, Shanghai, 200032, China
- Shanghai Key Laboratory of Acupuncture Mechanism and Acupoint Function, Shanghai Institute of Acupuncture and Moxibustion, Fudan University, Shanghai, 200433, China
| | - Rui Huang
- Department of Integrative Medicine and Neurobiology, School of Basic Medical Sciences, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science , Fudan University, Shanghai, 200032, China
- Shanghai Key Laboratory of Acupuncture Mechanism and Acupoint Function, Shanghai Institute of Acupuncture and Moxibustion, Fudan University, Shanghai, 200433, China
| | - Wei Hu
- Department of Integrative Medicine and Neurobiology, School of Basic Medical Sciences, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science , Fudan University, Shanghai, 200032, China.
- Shanghai Key Laboratory of Acupuncture Mechanism and Acupoint Function, Shanghai Institute of Acupuncture and Moxibustion, Fudan University, Shanghai, 200433, China.
| | - Yi Feng
- Department of Integrative Medicine and Neurobiology, School of Basic Medical Sciences, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science , Fudan University, Shanghai, 200032, China.
- Shanghai Key Laboratory of Acupuncture Mechanism and Acupoint Function, Shanghai Institute of Acupuncture and Moxibustion, Fudan University, Shanghai, 200433, China.
| |
Collapse
|
5
|
Yao J, Yao W, Zhu J, Liu Y, Liu J, Ji Y, Ni X, Mu W, Yan B. Targeting tRNA-Derived Non-Coding RNA Alleviates Diabetes-Induced Visual Impairment through Protecting Retinal Neurovascular Unit. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2411042. [PMID: 39513253 PMCID: PMC11714213 DOI: 10.1002/advs.202411042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/09/2024] [Indexed: 11/15/2024]
Abstract
Diabetes is a major risk factor for compromised visual health, leading to retinal vasculopathy and neuropathy, both of which are hallmarks of neurovascular unit dysfunction. Despite the critical impact of diabetic retinopathy, the precise mechanism underlying neurovascular coupling and effective strategies to suppress neurovascular dysfunction remain unclear. In this study, the up-regulation of a tRNA-derived stress-induced RNA, 5'tiRNA-His-GTG, in response to diabetic stress is revealed. 5'tiRNA-His-GTG directly regulates Müller glia action and indirectly alters endothelial angiogenic effects and retinal ganglion cell (RGC) survival in vitro. Downregulation of 5'tiRNA-His-GTG alleviates diabetes-induced retinal neurovascular dysfunction, characterized by reduced retinal vascular dysfunction, decreased retinal neurodegeneration, and improved visually-guided behaviors in vivo. Mechanistically, 5'tiRNA-His-GTG acts as a key regulator of retinal neurovascular dysfunction, primarily by modulating arachidonic acid (AA) metabolism via the CYPs pathway. The 5'tiRNA-His-GTG-CYP2E1-19(S)-HETE signaling axis is identified as a key driver of retinal neurovascular dysfunction. Thus, targeting 5'tiRNA-His-GTG presents a promising therapeutic strategy for treating vasculopathy and neuropathy associated with diabetes mellitus. Modulating this novel signaling pathway can open up new avenues for intervention in diabetic retinopathy and its related complications.
Collapse
Affiliation(s)
- Jin Yao
- The Affiliated Eye HospitalNanjing Medical UniversityNanjing210000China
| | - Wen Yao
- The Affiliated Eye HospitalNanjing Medical UniversityNanjing210000China
| | - Jun‐Ya Zhu
- The Affiliated Eye HospitalNanjing Medical UniversityNanjing210000China
- School of MedicineSoutheast UniversityNanjing210009China
| | - Yan Liu
- The Affiliated Eye HospitalNanjing Medical UniversityNanjing210000China
| | - Jin‐Hong Liu
- The Affiliated Eye HospitalNanjing Medical UniversityNanjing210000China
| | - Yu‐Ke Ji
- The Affiliated Eye HospitalNanjing Medical UniversityNanjing210000China
| | - Xi‐Shen Ni
- The Affiliated Eye HospitalNanjing Medical UniversityNanjing210000China
| | - Wan Mu
- Department of OphthalmologyShanghai General HospitalShanghai Jiao Tong University School of MedicineShanghai200080China
- Eye Institute and Department of OphthalmologyEye and ENT HospitalFudan UniversityShanghai200031China
| | - Biao Yan
- Department of OphthalmologyShanghai General HospitalShanghai Jiao Tong University School of MedicineShanghai200080China
| |
Collapse
|
6
|
Huo D, Bi XY, Zeng JL, Dai DM, Dong XL. Drugs targeting TGF-β/Notch interaction attenuate hypertrophic scar formation by optic atrophy 1-mediated mitochondrial fusion. Mol Cell Biochem 2024; 479:3049-3061. [PMID: 38158493 DOI: 10.1007/s11010-023-04912-y] [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: 06/30/2023] [Accepted: 11/29/2023] [Indexed: 01/03/2024]
Abstract
Hypertrophic scar (HS) formation is a cutaneous fibroproliferative disease that occurs after skin injuries and results in severe functional and esthetic disability. To date, few drugs have shown satisfactory outcomes for the treatment of HS formation. Transforming growth factor-beta (TGF-β)/Notch interaction via small mothers against decapentaplegic 3 (Smad3) could facilitate HS formation; therefore, targeting TGF-β/ Notch interaction via Smad3 is a potential therapeutic strategy to attenuate HS formation. In addition, optic atrophy 1 (OPA1)-mediated mitochondrial fusion contributes to fibroblast proliferation, and TGF-β/Smad3 axis and the Notch1 pathway facilitate OPA1-mediated mitochondrial fusion. Thus, the aim of this study was to investigate whether drugs targeting TGF-β/Notch interaction via Smad3 suppressed fibroblast proliferation to attenuate HS formation through OPA1-mediated mitochondrial fusion. We found that the TGF-β pathway, Notch pathway, and TGF-β/Notch interaction via Smad3 were inhibited by pirfenidone, the gamma- secretase inhibitor DAPT, and SIS3 in human keloid fibroblasts (HKF) and an HS rat model, respectively. Protein interaction was detected by co-immunoprecipitation, and mitochondrial morphology was determined by electron microscopy. Our results indicated that pirfenidone, DAPT, and SIS3 suppressed the proliferation of HKFs and attenuated HS formation in the HS rat model by inhibiting TGF-β/Notch interaction via Smad3. Moreover, pirfenidone, DAPT, and SIS3 hindered OPA1-mediated mitochondrial fusion through inhibiting TGF-β/Notch interaction, thereby suppressing the proliferation of HS fibroblasts and HS formation. In summary, these findings investigating the effects of drugs targeting TGF-β/Notch interaction on HS formation might lead to novel drugs for the treatment of HS formation.
Collapse
Affiliation(s)
- Da Huo
- Department of Plastic and Aesthetic, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, 830011, Xinjiang, People's Republic of China
| | - Xin-Yu Bi
- Department of Rehabilitation Medicine, The First Affiliated Hospital of China Medical University, Shenyang, 110001, Liaoning Province, People's Republic of China
| | - Jun-Ling Zeng
- Laboratory Animal Research Center of Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong, People's Republic of China
| | - Da-Mao Dai
- Department of Plastic and Cosmetic Surgery, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, 518001, Guangdong, People's Republic of China.
| | - Xiang-Lin Dong
- Department of Plastic and Aesthetic, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, 830011, Xinjiang, People's Republic of China.
| |
Collapse
|
7
|
Prieto-López L, Pereiro X, Vecino E. The mechanics of the retina: Müller glia role on retinal extracellular matrix and modelling. Front Med (Lausanne) 2024; 11:1393057. [PMID: 39296899 PMCID: PMC11410058 DOI: 10.3389/fmed.2024.1393057] [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: 03/04/2024] [Accepted: 08/13/2024] [Indexed: 09/21/2024] Open
Abstract
The retina is a highly heterogeneous tissue, both cell-wise but also regarding its extracellular matrix (ECM). The stiffness of the ECM is pivotal in retinal development and maturation and has also been associated with the onset and/or progression of numerous retinal pathologies, such as glaucoma, proliferative vitreoretinopathy (PVR), age-related macular degeneration (AMD), epiretinal membrane (ERM) formation or uveitis. Nonetheless, much remains unknown about the biomechanical milieu of the retina, and specifically the role that Müller glia play as principal mechanosensors and major producers of ECM constituents. So far, new approaches need to be developed to further the knowledge in the field of retinal mechanobiology for ECM-target applications to arise. In this review, we focus on the involvement of Müller glia in shaping and altering the retinal ECM under both physiological and pathological conditions and look into various biomaterial options to more accurately replicate the impact of matrix stiffness in vitro.
Collapse
Affiliation(s)
- Laura Prieto-López
- Experimental Ophthalmo-Biology Group, Department of Cell Biology and Histology, University of Basque Country UPV/EHU, Leioa, Spain
| | - Xandra Pereiro
- Experimental Ophthalmo-Biology Group, Department of Cell Biology and Histology, University of Basque Country UPV/EHU, Leioa, Spain
- Begiker-Ophthalmology Research Group, BioCruces Health Research Institute, Cruces Hospital, Barakaldo, Spain
| | - Elena Vecino
- Experimental Ophthalmo-Biology Group, Department of Cell Biology and Histology, University of Basque Country UPV/EHU, Leioa, Spain
- Begiker-Ophthalmology Research Group, BioCruces Health Research Institute, Cruces Hospital, Barakaldo, Spain
| |
Collapse
|
8
|
Zhou Y, Zhou W, Rao Y, He J, Huang Y, Zhao P, Li J. Dysregulated energy and protein homeostasis and the loss of GABAergic amacrine cells in aging retina. Exp Eye Res 2024; 245:109985. [PMID: 38945518 DOI: 10.1016/j.exer.2024.109985] [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: 02/26/2024] [Revised: 05/30/2024] [Accepted: 06/27/2024] [Indexed: 07/02/2024]
Abstract
Aging is a major risk factor for the development or the worsening of retinal degenerative conditions. The intricate network of the neural retina determined that the retinal aging is a complicated process. The aim of this study is to delineate the transcriptomic changes of major retinal neurons during aging in C57BL/6 mice at single-cell level. We analyzed the transcriptional profiles of the photoreceptor, bipolar, amacrine, and Müller glial cells of 1.5-2 and 24-30 months old mice using single-cell RNA sequencing technique. We selectively confirmed the differences in gene expression using immunofluorescence staining and RNA in situ hybridization analysis. We found that each retinal cell type had unique changes upon aging. However, they all showed signs of dysregulated glucose and energy metabolism, and perturbed proteostasis. In particular, old Müller glia exhibited the most profound changes, including the upregulation of cell metabolism, stress-responses, antigen-presentation and immune responses and metal ion homeostasis. The dysregulated gliogenesis and differentiation was confirmed by the presence of Müller glia expressing rod-specific genes in the inner nuclear layer and the outer plexiform layer of the old retina. We further pinpointed the specific loss of GABAergic amacrine cells in old retina. Our study emphasized changes of amacrine and Müller glia during retinal aging, provided resources for further research on the molecular and cellular regulatory mechanisms underlying aging-associated retinal deterioration.
Collapse
Affiliation(s)
- Yutong Zhou
- Department of Ophthalmology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
| | - Wenchuan Zhou
- Department of Ophthalmology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
| | - Yuqing Rao
- Department of Ophthalmology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
| | - Jincan He
- Department of Ophthalmology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
| | - Yue Huang
- Department of Ophthalmology, Chongming Hospital Affiliated to Shanghai University of Medicine and Health Sciences, Shanghai, 202150, China
| | - Peiquan Zhao
- Department of Ophthalmology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China.
| | - Jing Li
- Department of Ophthalmology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China.
| |
Collapse
|
9
|
Hung JH, Tsai PH, Aala WJF, Chen CC, Chiou SH, Wong TW, Tsai KJ, Hsu SM, Wu LW. TIMP3/Wnt axis regulates gliosis of Müller glia. Biochim Biophys Acta Mol Basis Dis 2024; 1870:167087. [PMID: 38369214 DOI: 10.1016/j.bbadis.2024.167087] [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: 11/01/2023] [Revised: 02/11/2024] [Accepted: 02/15/2024] [Indexed: 02/20/2024]
Abstract
BACKGROUND Previous studies have confirmed the expression of tissue inhibitor of metalloproteinase-3 (TIMP3) in Müller glia (MG). However, the role of TIMP3 in MG remains unknown. METHODS A mouse model of laser-induced retinal damage and gliosis was generated using wild-type C57BL/6 mice. TIMP3 and associated proteins were detected using Western blotting and immunofluorescence microscopy. RNA sequencing (GSE132140) of mouse laser-induced gliosis was utilized for pathway analysis. TIMP3 overexpression was induced in human MG. Human vitreous samples were obtained from patients with proliferative diabetic retinopathy (PDR) and healthy controls for protein analysis. RESULTS TIMP3 levels increased in mouse eyes after laser damage. Morphology and spatial location of TIMP3 indicated its presence in MG. TIMP3-overexpressing MG showed increased cellular proliferation, migration, and cell nuclei size, suggesting TIMP3-induced gliosis for retinal repair. Glial fibrillary acidic protein (GFAP) and vimentin levels were elevated in TIMP3-overexpressing MG and laser-damaged mouse retinas. RNA sequencing and Western blotting suggested a role for β-catenin in mediating TIMP3 effects on the retina. Human vitreous samples from patients with PDR showed a positive correlation between TIMP3 and GFAP levels, both of which were elevated in patients with PDR. CONCLUSIONS TIMP3 is associated with MG gliosis to enhance the repair ability of damaged retinas and is mediated by the canonical Wnt/β-catenin. Changes in TIMP3 could potentially be used to control gliosis in a range of retinal diseases However, given the multifaceted nature of TIMP3, care must be taken when developing treatments that aim solely to boost the function of TIMP3. FUNDING National Cheng Kung University Hospital, Taiwan (NCKUH-10604009 and NCKUH-11202007); the Ministry of Science and Technology (MOST 110-2314-B-006-086-MY3).
Collapse
Affiliation(s)
- Jia-Horung Hung
- Institute of Clinical Medicine, College of Medicine, National Cheng Kung University, Tainan, Taiwan; Department of Ophthalmology, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Ping-Hsing Tsai
- Department of Medical Research, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Wilson Jr F Aala
- Institute of Clinical Medicine, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Chao-Chung Chen
- Department of Ophthalmology, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Shih-Hwa Chiou
- Department of Medical Research, Taipei Veterans General Hospital, Taipei, Taiwan; Institute of Pharmacology, School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Tak-Wah Wong
- Department of Dermatology, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan; Department of Biochemistry and Molecular Biology, College of Medicine, National Cheng Kung University, Tainan, Taiwan; Center of Applied Nanomedicine, National Cheng Kung University, Tainan, Taiwan
| | - Kuen-Jer Tsai
- Institute of Clinical Medicine, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Sheng-Min Hsu
- Department of Ophthalmology, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan.
| | - Li-Wha Wu
- Institute of Molecular Medicine, College of Medicine, National Cheng Kung University, Tainan, Taiwan; Department of Medical Laboratory Science and Biotechnology, Kaohsiung Medical University, Kaohsiung, Taiwan.
| |
Collapse
|
10
|
Conedera FM, Kokona D, Zinkernagel MS, Stein JV, Lin CP, Alt C, Enzmann V. Macrophages coordinate immune response to laser-induced injury via extracellular traps. J Neuroinflammation 2024; 21:68. [PMID: 38500151 PMCID: PMC10949579 DOI: 10.1186/s12974-024-03064-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Accepted: 03/13/2024] [Indexed: 03/20/2024] Open
Abstract
BACKGROUND Retinal degeneration results from disruptions in retinal homeostasis due to injury, disease, or aging and triggers peripheral leukocyte infiltration. Effective immune responses rely on coordinated actions of resident microglia and recruited macrophages, critical for tissue remodeling and repair. However, these phagocytes also contribute to chronic inflammation in degenerated retinas, yet the precise coordination of immune response to retinal damage remains elusive. Recent investigations have demonstrated that phagocytic cells can produce extracellular traps (ETs), which are a source of self-antigens that alter the immune response, which can potentially lead to tissue injury. METHODS Innovations in experimental systems facilitate real-time exploration of immune cell interactions and dynamic responses. We integrated in vivo imaging with ultrastructural analysis, transcriptomics, pharmacological treatments, and knockout mice to elucidate the role of phagocytes and their modulation of the local inflammatory response through extracellular traps (ETs). Deciphering these mechanisms is essential for developing novel and enhanced immunotherapeutic approaches that can redirect a specific maladaptive immune response towards favorable wound healing in the retina. RESULTS Our findings underscore the pivotal role of innate immune cells, especially macrophages/monocytes, in regulating retinal repair and inflammation. The absence of neutrophil and macrophage infiltration aids parenchymal integrity restoration, while their depletion, particularly macrophages/monocytes, impedes vascular recovery. We demonstrate that macrophages/monocytes, when recruited in the retina, release chromatin and granular proteins, forming ETs. Furthermore, the pharmacological inhibition of ETosis support retinal and vascular repair, surpassing the effects of blocking innate immune cell recruitment. Simultaneously, the absence of ETosis reshapes the inflammatory response, causing neutrophils, helper, and cytotoxic T-cells to be restricted primarily in the superficial capillary plexus instead of reaching the damaged photoreceptor layer. CONCLUSIONS Our data offer novel insights into innate immunity's role in responding to retinal damage and potentially help developing innovative immunotherapeutic approaches that can shift the immune response from maladaptive to beneficial for retinal regeneration.
Collapse
Affiliation(s)
- Federica M Conedera
- Department of Oncology, Microbiology and Immunology, University of Fribourg, Fribourg, Switzerland.
- Department of Ophthalmology, Bern University Hospital and Department of BioMedical Research, University of Bern, Bern, Switzerland.
| | - Despina Kokona
- Department of Ophthalmology, Bern University Hospital and Department of BioMedical Research, University of Bern, Bern, Switzerland
| | - Martin S Zinkernagel
- Department of Ophthalmology, Bern University Hospital and Department of BioMedical Research, University of Bern, Bern, Switzerland
| | - Jens V Stein
- Department of Oncology, Microbiology and Immunology, University of Fribourg, Fribourg, Switzerland
| | - Charles P Lin
- Center for Systems Biology and Wellman Center for Photomedicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Clemens Alt
- Center for Systems Biology and Wellman Center for Photomedicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Volker Enzmann
- Department of Ophthalmology, Bern University Hospital and Department of BioMedical Research, University of Bern, Bern, Switzerland
| |
Collapse
|
11
|
Wu X, Qin B, Cheng R, Zhou R, Wang X, Zhang Z, Mao X, Xie Z, Chen M, Jiang L, Xie P, Ji J, Zhang W, Yuan S, Hu Z, Liu Q. Angiogenic and Fibrogenic Dual-effect of Gremlin1 on Proliferative Diabetic Retinopathy. Int J Biol Sci 2024; 20:897-915. [PMID: 38250154 PMCID: PMC10797694 DOI: 10.7150/ijbs.85735] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2023] [Accepted: 10/24/2023] [Indexed: 01/23/2024] Open
Abstract
Ocular angiogenic diseases, such as proliferative diabetic retinopathy (PDR), are often characterized by pathological new vessels and fibrosis formation. Anti-vascular endothelial growth factor (VEGF) therapy, despite of its efficiency to inhibit new vessels, has limitations, including drug resistance and retinal fibrosis. Here, we identified that Gremlin1, a novel angiogenesis and fibrosis inducer, was secreted from Müller glial cells, and its expression increased in the vitreous fluid from patients with PDR. Mechanistically, Gremlin1 triggered angiogenesis by promoting endothelial-mesenchymal transition via the EGFR/RhoA/ROCK pathway. In addition, Gremlin1 activated microglia to present profibrotic and fibrogenic properties. Further, anti-Gremlin1 antibody inhibited ocular angiogenesis and microglia fibrosis in mouse models. Collectively, Gremlin1 could be a potential therapeutic target in the treatment of ocular angiogenic diseases.
Collapse
Affiliation(s)
- Xinjing Wu
- Department of Ophthalmology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Bing Qin
- Department of Ophthalmology, The Affiliated Suqian First People's Hospital of Nanjing Medical University, Suqian 223800, China
| | - Ruiwen Cheng
- Department of Ophthalmology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Ru Zhou
- Department of Ophthalmology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
- Department of Ophthalmology, People's Hospital of Yangzhong City, Yangzhong 212200, China
| | - Xingxing Wang
- Department of Ophthalmology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Zhengyu Zhang
- Department of Ophthalmology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Xiying Mao
- Department of Ophthalmology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Zhan Xie
- Department of Ophthalmology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Mingkang Chen
- Department of Ophthalmology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Lin Jiang
- Department of Endocrinology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Ping Xie
- Department of Ophthalmology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Jiangdong Ji
- Department of Ophthalmology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Weiwei Zhang
- Department of Ophthalmology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Songtao Yuan
- Department of Ophthalmology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Zizhong Hu
- Department of Ophthalmology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Qinghuai Liu
- Department of Ophthalmology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| |
Collapse
|
12
|
Han H, Tang L, Li Y, Li Y, Bi M, Wang J, Wang F, Wang L, Mao J. A multifunctional surgical suture with electroactivity assisted by oligochitosan/gelatin-tannic acid for promoting skin wound healing and controlling scar proliferation. Carbohydr Polym 2023; 320:121236. [PMID: 37659821 DOI: 10.1016/j.carbpol.2023.121236] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Revised: 07/23/2023] [Accepted: 07/25/2023] [Indexed: 09/04/2023]
Abstract
Surgical wound closure is accomplished most frequently with sutures, optimally proceeding rapidly and without complication. However, surgical sutures can trigger foreign body reactions and incite abnormal collagen deposition. Sustained inflammation can result in abnormal wound healing with hypertrophic scar formation. Therefore, evolution of suture material to inhibit inflammation and scar formation is of great clinical significance. In the present study, commercial 3-0 PPDO [poly(p-dioxanone)] suture was used as the base material and modified by adding two layers: a drug-loaded layer and an electroactive layer. The former layer was curcumin (Cur) encapsulated by PLGA [poly (lactic-co-glycolic acid)] and the latter layer was composed of oligochitosan-gelatin/tannic acid/polypyrrole (OCS-GE/TA/PPy). The multifunctional sutures, named S@LC@CGTP, had desirable sustained-drug release properties in vitro where Cur could be released for 8 days due to the action of PLGA. The three-dimensional network structure of OCS-GE/TA ensured S@LC@CGTP against surface cracking and maintained electrical. Furthermore, using an in vivo experiment, S@LC@CGTP could attenuate inflammation and promote scar-free wound healing according to suppression of infiltrating inflammatory cells, down-regulation of TGF-β1 and collagen type I expression, and improved collagen arrangement. Cumulatively, we indicated that S@LC@CGTP suture material has great potential to facilitate optimal, nearly scarless healing of surgical incisions.
Collapse
Affiliation(s)
- Hui Han
- Thyroid Surgery Department, General Surgery Center, The First Hospital of Jilin University, Changchun, Jilin 130021, China
| | - Liqin Tang
- Key Laboratory of Textile Science & Technology, Ministry of Education, Donghua University, Shanghai 201620, China; Key Laboratory of Textile Industry for Biomedical Textile Materials and Technology, Donghua University, Shanghai 201620, China
| | - Yan Li
- Key Laboratory of Textile Science & Technology, Ministry of Education, Donghua University, Shanghai 201620, China; Key Laboratory of Textile Industry for Biomedical Textile Materials and Technology, Donghua University, Shanghai 201620, China
| | - Yong Li
- Thyroid Surgery Department, General Surgery Center, The First Hospital of Jilin University, Changchun, Jilin 130021, China
| | - Ming Bi
- General department, Hospital of Stomatology, Jilin University, Changchun, Jilin 130021, China.
| | - Jun Wang
- Key Laboratory of Textile Science & Technology, Ministry of Education, Donghua University, Shanghai 201620, China
| | - Fujun Wang
- Key Laboratory of Textile Science & Technology, Ministry of Education, Donghua University, Shanghai 201620, China; Key Laboratory of Textile Industry for Biomedical Textile Materials and Technology, Donghua University, Shanghai 201620, China; Shanghai Frontiers Science Center of Advanced Textiles, Donghua University, Shanghai 201620, China
| | - Lu Wang
- Key Laboratory of Textile Science & Technology, Ministry of Education, Donghua University, Shanghai 201620, China; Key Laboratory of Textile Industry for Biomedical Textile Materials and Technology, Donghua University, Shanghai 201620, China; Shanghai Frontiers Science Center of Advanced Textiles, Donghua University, Shanghai 201620, China
| | - Jifu Mao
- Key Laboratory of Textile Science & Technology, Ministry of Education, Donghua University, Shanghai 201620, China; Key Laboratory of Textile Industry for Biomedical Textile Materials and Technology, Donghua University, Shanghai 201620, China; Shanghai Frontiers Science Center of Advanced Textiles, Donghua University, Shanghai 201620, China.
| |
Collapse
|
13
|
Conedera FM, Runnels JM, Stein JV, Alt C, Enzmann V, Lin CP. Assessing the role of T cells in response to retinal injury to uncover new therapeutic targets for the treatment of retinal degeneration. J Neuroinflammation 2023; 20:206. [PMID: 37689689 PMCID: PMC10492418 DOI: 10.1186/s12974-023-02867-x] [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: 03/03/2023] [Accepted: 07/31/2023] [Indexed: 09/11/2023] Open
Abstract
BACKGROUND Retinal degeneration is a disease affecting the eye, which is an immune-privileged site because of its anatomical and physiological properties. Alterations in retinal homeostasis-because of injury, disease, or aging-initiate inflammatory cascades, where peripheral leukocytes (PL) infiltrate the parenchyma, leading to retinal degeneration. So far, research on PL's role in retinal degeneration was limited to observing a few cell types at specific times or sectioning the tissue. This restricted our understanding of immune cell interactions and response duration. METHODS In vivo microscopy in preclinical mouse models can overcome these limitations enabling the spatio-temporal characterization of PL dynamics. Through in vivo imaging, we assessed structural and fluorescence changes in response to a focal injury at a defined location over time. We also utilized minimally invasive techniques, pharmacological interventions, and knockout (KO) mice to determine the role of PL in local inflammation. Furthermore, we investigated PL abundance and localization during retinal degeneration in human eyes by histological analysis to assess to which extent our preclinical study translates to human retinal degeneration. RESULTS We demonstrate that PL, especially T cells, play a detrimental role during retinal injury response. In mice, we observed the recruitment of helper and cytotoxic T cells in the parenchyma post-injury, and T cells also resided in the macula and peripheral retina in pathological conditions in humans. Additionally, we found that the pharmacological PL reduction and genetic depletion of T-cells reduced injured areas in murine retinas and rescued the blood-retina barrier (BRB) integrity. Both conditions promoted morphological changes of Cx3cr1+ cells, including microglial cells, toward an amoeboid phenotype during injury response. Interestingly, selective depletion of CD8+ T cells accelerated recovery of the BRB compared to broader depletions. After anti-CD8 treatment, the retinal function improved, concomitant to a beneficial immune response. CONCLUSIONS Our data provide novel insights into the adaptive immune response to retinal injury in mice and human retinal degeneration. Such information is fundamental to understanding retinal disorders and developing therapeutics to modulate immune responses to retinal degeneration safely.
Collapse
Affiliation(s)
- Federica M Conedera
- Department of Oncology, Microbiology and Immunology, University of Fribourg, Fribourg, Switzerland
- Department of Ophthalmology, Bern University Hospital, Bern, Switzerland
| | - Judith M Runnels
- Center for Systems Biology and Wellman Center for Photomedicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Jens V Stein
- Department of Oncology, Microbiology and Immunology, University of Fribourg, Fribourg, Switzerland
| | - Clemens Alt
- Center for Systems Biology and Wellman Center for Photomedicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Volker Enzmann
- Department of Ophthalmology, Bern University Hospital, Bern, Switzerland.
- Department of BioMedical Research, University of Bern, Bern, Switzerland.
| | - Charles P Lin
- Center for Systems Biology and Wellman Center for Photomedicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| |
Collapse
|
14
|
Liu C, Nikain C, Li YM. γ-Secretase fanning the fire of innate immunity. Biochem Soc Trans 2023; 51:1597-1610. [PMID: 37449907 PMCID: PMC11212119 DOI: 10.1042/bst20221445] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Revised: 06/29/2023] [Accepted: 07/03/2023] [Indexed: 07/18/2023]
Abstract
Innate immunity is the first line of defense against pathogens, alerting the individual cell and surrounding area to respond to this potential invasion. γ-secretase is a transmembrane protease complex that plays an intricate role in nearly every stage of this innate immune response. Through regulation of pattern recognition receptors (PRR) such as TREM2 and RAGE γ-secretase can modulate pathogen recognition. γ-secretase can act on cytokine receptors such as IFNαR2 and CSF1R to dampen their signaling capacity. While γ-secretase-mediated regulated intramembrane proteolysis (RIP) can further moderate innate immune responses through downstream signaling pathways. Furthermore, γ-secretase has also been shown to be regulated by the innate immune system through cytokine signaling and γ-secretase modulatory proteins such as IFITM3 and Hif-1α. This review article gives an overview of how γ-secretase is implicated in innate immunity and the maintenance of its responses through potentially positive and negative feedback loops.
Collapse
Affiliation(s)
- Chenge Liu
- Chemical Biology Program, Memorial Sloan Kettering Cancer Center
- Programs of Pharmacology, Weill Graduate School of Medical Sciences of Cornell University
| | - Cyrus Nikain
- Chemical Biology Program, Memorial Sloan Kettering Cancer Center
- Programs of Pharmacology, Weill Graduate School of Medical Sciences of Cornell University
| | - Yue-Ming Li
- Chemical Biology Program, Memorial Sloan Kettering Cancer Center
- Programs of Pharmacology, Weill Graduate School of Medical Sciences of Cornell University
| |
Collapse
|
15
|
Xiao X, Liao Z, Zou J. Genetic and epigenetic regulators of retinal Müller glial cell reprogramming. ADVANCES IN OPHTHALMOLOGY PRACTICE AND RESEARCH 2023; 3:126-133. [PMID: 37846362 PMCID: PMC10577857 DOI: 10.1016/j.aopr.2023.05.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Revised: 05/18/2023] [Accepted: 05/29/2023] [Indexed: 10/18/2023]
Abstract
Background Retinal diseases characterized with irreversible loss of retinal nerve cells, such as optic atrophy and retinal degeneration, are the main causes of blindness. Current treatments for these diseases are very limited. An emerging treatment strategy is to induce the reprogramming of Müller glial cells to generate new retinal nerve cells, which could potentially restore vision. Main text Müller glial cells are the predominant glial cells in retinae and play multiple roles to maintain retinal homeostasis. In lower vertebrates, such as in zebrafish, Müller glial cells can undergo cell reprogramming to regenerate new retinal neurons in response to various damage factors, while in mammals, this ability is limited. Interestingly, with proper treatments, Müller glial cells can display the potential for regeneration of retinal neurons in mammalian retinae. Recent studies have revealed that dozens of genetic and epigenetic regulators play a vital role in inducing the reprogramming of Müller glial cells in vivo. This review summarizes these critical regulators for Müller glial cell reprogramming and highlights their differences between zebrafish and mammals. Conclusions A number of factors have been identified as the important regulators in Müller glial cell reprogramming. The early response of Müller glial cells upon acute retinal injury, such as the regulation in the exit from quiescent state, the initiation of reactive gliosis, and the re-entry of cell cycle of Müller glial cells, displays significant difference between mouse and zebrafish, which may be mediated by the diverse regulation of Notch and TGFβ (transforming growth factor-β) isoforms and different chromatin accessibility.
Collapse
Affiliation(s)
- Xueqi Xiao
- Zhejiang Provincial Key Laboratory for Water Environment and Marine Biological Resources Protection, College of Life and Environmental Science, Wenzhou University, Wenzhou, China
| | - Zhiyong Liao
- Zhejiang Provincial Key Laboratory for Water Environment and Marine Biological Resources Protection, College of Life and Environmental Science, Wenzhou University, Wenzhou, China
| | - Jian Zou
- Department of Ophthalmology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
- The Institute of Translational Medicine, Zhejiang University, Hangzhou, China
| |
Collapse
|
16
|
Krylov A, Yu S, Veen K, Newton A, Ye A, Qin H, He J, Jusuf PR. Heterogeneity in quiescent Müller glia in the uninjured zebrafish retina drive differential responses following photoreceptor ablation. Front Mol Neurosci 2023; 16:1087136. [PMID: 37575968 PMCID: PMC10413128 DOI: 10.3389/fnmol.2023.1087136] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Accepted: 06/23/2023] [Indexed: 08/15/2023] Open
Abstract
Introduction Loss of neurons in the neural retina is a leading cause of vision loss. While humans do not possess the capacity for retinal regeneration, zebrafish can achieve this through activation of resident Müller glia. Remarkably, despite the presence of Müller glia in humans and other mammalian vertebrates, these cells lack an intrinsic ability to contribute to regeneration. Upon activation, zebrafish Müller glia can adopt a stem cell-like state, undergo proliferation and generate new neurons. However, the underlying molecular mechanisms of this activation subsequent retinal regeneration remains unclear. Methods/Results To address this, we performed single-cell RNA sequencing (scRNA-seq) and report remarkable heterogeneity in gene expression within quiescent Müller glia across distinct dorsal, central and ventral retina pools of such cells. Next, we utilized a genetically driven, chemically inducible nitroreductase approach to study Müller glia activation following selective ablation of three distinct photoreceptor subtypes: long wavelength sensitive cones, short wavelength sensitive cones, and rods. There, our data revealed that a region-specific bias in activation of Müller glia exists in the zebrafish retina, and this is independent of the distribution of the ablated cell type across retinal regions. Notably, gene ontology analysis revealed that injury-responsive dorsal and central Müller glia express genes related to dorsal/ventral pattern formation, growth factor activity, and regulation of developmental process. Through scRNA-seq analysis, we identify a shared genetic program underlying initial Müller glia activation and cell cycle entry, followed by differences that drive the fate of regenerating neurons. We observed an initial expression of AP-1 and injury-responsive transcription factors, followed by genes involved in Notch signaling, ribosome biogenesis and gliogenesis, and finally expression of cell cycle, chromatin remodeling and microtubule-associated genes. Discussion Taken together, our findings document the regional specificity of gene expression within quiescent Müller glia and demonstrate unique Müller glia activation and regeneration features following neural ablation. These findings will improve our understanding of the molecular pathways relevant to neural regeneration in the retina.
Collapse
Affiliation(s)
- Aaron Krylov
- School of BioSciences, University of Melbourne, Parkville, VIC, Australia
| | - Shuguang Yu
- State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Institute of Neuroscience, Chinese Academy of Sciences, Shanghai, China
| | - Kellie Veen
- School of BioSciences, University of Melbourne, Parkville, VIC, Australia
| | - Axel Newton
- School of BioSciences, University of Melbourne, Parkville, VIC, Australia
| | - Aojun Ye
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
| | - Huiwen Qin
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
| | - Jie He
- State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Institute of Neuroscience, Chinese Academy of Sciences, Shanghai, China
| | - Patricia R. Jusuf
- School of BioSciences, University of Melbourne, Parkville, VIC, Australia
| |
Collapse
|
17
|
Jahnke L, Zandi S, Elhelbawi A, Conedera FM, Enzmann V. Characterization of Macroglia Response during Tissue Repair in a Laser-Induced Model of Retinal Degeneration. Int J Mol Sci 2023; 24:ijms24119172. [PMID: 37298126 DOI: 10.3390/ijms24119172] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2023] [Revised: 05/18/2023] [Accepted: 05/19/2023] [Indexed: 06/12/2023] Open
Abstract
Reactive gliosis is a hallmark of chronic degenerative diseases of the retina. As gliosis involves macroglia, we investigated their gliotic response to determine the role of S100β and intermediate filaments (IFs) GFAP, vimentin, and nestin during tissue repair in a laser-induced model of retinal degeneration. We validated the results with human retinal donor samples. Experiments were performed in zebrafish and mice using an argon laser (532 nm) to induce focal lesions in the outer retina. At different time points following injury induction, the kinetics of retinal degeneration and regeneration were assessed using hematoxylin and eosin staining (H&E). Immunofluorescence was performed to evaluate Müller cell (GS) and astrocyte (GFAP) injury response and to distinguish between both cell types. Additionally, staining was performed in human retinal sections containing drusen. Focal laser treatment elevated the expression of gliotic markers in the area of the damage, which was associated with increased expression of S100β, GFAP, vimentin, and nestin in mice and humans. In zebrafish, we detected S100β at the first time point, but not GFAP or nestin. Double-positive cells with the selected glia markers were detected in all models. However, in zebrafish, no double-positive GFAP/GS cells were found on days 10 and 17, nor were S100β/GS double-positive cells found on day 12. Macroglia cells showed a different pattern in the expression of IFs in degenerative and regenerative models. In particular, S100β may prove to be a target for suppressing chronic gliosis in retinal degeneration.
Collapse
Affiliation(s)
- Laura Jahnke
- Department of Ophthalmology, Inselspital, Bern University Hospital, University of Bern, 3010 Bern, Switzerland
- Department of BioMedical Research, University of Bern, 3008 Bern, Switzerland
- Graduate School for Cellular and Biomedical Sciences, University of Bern, 3012 Bern, Switzerland
| | - Souska Zandi
- Department of Ophthalmology, Inselspital, Bern University Hospital, University of Bern, 3010 Bern, Switzerland
- Department of BioMedical Research, University of Bern, 3008 Bern, Switzerland
| | - Ahmed Elhelbawi
- Graduate School for Cellular and Biomedical Sciences, University of Bern, 3012 Bern, Switzerland
- Department of Chemistry, Biochemistry and Pharmaceutical Sciences, University of Bern, 3012 Bern, Switzerland
| | | | - Volker Enzmann
- Department of Ophthalmology, Inselspital, Bern University Hospital, University of Bern, 3010 Bern, Switzerland
- Department of BioMedical Research, University of Bern, 3008 Bern, Switzerland
| |
Collapse
|
18
|
Xu D, Gao C, Cao Y, Xiao B. HOXC8 alleviates high glucose-triggered damage of trophoblast cells during gestational diabetes mellitus via activating TGFβ1-mediated Notch1 pathway. Hum Cell 2023; 36:195-208. [PMID: 36308681 DOI: 10.1007/s13577-022-00816-z] [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: 04/20/2022] [Accepted: 10/18/2022] [Indexed: 01/07/2023]
Abstract
Gestational diabetes mellitus (GDM) is an increasingly frequent disease occurred during pregnancy. HOXC8 has been disclosed to take part in the regulation of cancers. Additionally, the HOXC8 expression was dramatically decreased in the placenta of pre-eclampsia patients, but its expression and function have not been investigated in GDM. In this work, it was demonstrated that the mRNA and protein expression of HOXC8 was lower in GDM placenta tissues and GDM cell model. In addition, HOXC8 facilitated trophoblast cell proliferation and weakened trophoblast cell mitochondrial apoptosis. HOXC8 enhanced trophoblast cell migration and angiogenesis. Moreover, HOXC8 activated the TGFβ1-mediated Notch1 signaling pathway. Results showed that the mRNA and protein expressions of TGFβ1 and Notch1 were both lower in the GDM group than that in the NP group. Besides, there were positive correlations among HOXC8, TGFβ1 and Notch1. Inhibition of TGFβ1 (SB202190 treatment) reversed the effects of HOXC8 on trophoblast cells through modulating cell proliferation, mitochondrial apoptosis, migration and angiogenesis. At last, through in vivo experiments, it was identified that HOXC8 relieved GDM symptoms in vivo. In conclusion, HOXC8 alleviated HG-stimulated damage of trophoblast cells during GDM through activating TGFβ1-mediated Notch1 pathway. This discovery may provide a novel and useful bio-target for GDM treatment.
Collapse
Affiliation(s)
- Dan Xu
- Department of Obstetrics, The Yancheng Clinical College of Xuzhou Medical University, The First people's Hospital of Yancheng, Yancheng, 224001, Jiangsu, PR China
| | - Chengzhen Gao
- Department of Obstetrics, The Yancheng Clinical College of Xuzhou Medical University, The First people's Hospital of Yancheng, Yancheng, 224001, Jiangsu, PR China
| | - Yuanyuan Cao
- Department of Obstetrics, The Yancheng Clinical College of Xuzhou Medical University, The First people's Hospital of Yancheng, Yancheng, 224001, Jiangsu, PR China
| | - Biru Xiao
- Department of Obstetrics, The First Affiliated Hospital of Wenzhou Medical University, Nanbaixiang, Ouhai District, Wenzhou, 325000, Zhejiang, PR China.
| |
Collapse
|
19
|
Enzmann V, Conedera F. Regenerative capacity of Müller cells and their modulation as a tool to treat retinal degenerations. Neural Regen Res 2023; 18:139-140. [PMID: 35799533 PMCID: PMC9241422 DOI: 10.4103/1673-5374.340408] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
|
20
|
Chen Y, Tian B. IFN-γ promotes the development of systemic lupus erythematosus through the IFNGR1/2-PSTAT1-TBX21 signaling axis. Am J Transl Res 2022; 14:6874-6888. [PMID: 36398225 PMCID: PMC9641460] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Accepted: 08/18/2022] [Indexed: 06/16/2023]
Abstract
BACKGROUND Systemic lupus erythematosus (SLE) is a chronic disease that causes inflammation in cartilage and the lining of blood vessels. Emerging evidence implicates IFN-γ as a major effector molecule in SLE during both active and stable stages. Here, we investigated the effects of IFN-γ on cytokines that play an autoimmune disease-promoting role and Th1-versus-Th2 and B cell dualism in SLE patients and mouse models of SLE. METHODS The levels of pro-inflammatory factors CXCL11, IFN-γ, IL-1β and IL-4, and immune complexes IgG, anti-dsDNA and anti-RNP were assessed through enzyme-linked immunosorbent assays (ELISA). Flow cytometry was performed to measure Th1, Th2 and B cell counts and IFNGR1, IFNGR2, pSTAT1 and TBX21 expression. The pathology of renal tissue from mouse SLE models was investigated through Hematoxylin eosin (H&E) staining. The levels of IgG, anti-dsDNA and anti-RNP were determined through immunofluorescence (IF) assays. RESULTS Skin damage was observed in SLE patients in both active and stable stages. ELISA analysis showed that SLE patients displayed higher levels of pro-inflammatory factors (CXCL11, IFN-γ, IL-1β and IL-4) and immune complexes (IgG, anti-dsDNA and anti-RNP). The percentage of Th1 and B cells was increased in blood samples from SLE patients with skin lesions (SL) or lupus nephritis (LN). The percentage of Th2 cells among the groups were comparable. Higher levels of IFNGR1, IFNGR2, pSTAT1 and TBX21 were observed in Th1 but not Th2 cells. In SLE mouse models, H&E staining revealed fewer immune complexes in glomerular endothelial cells and decreased hyaline thrombus in the capillary lumen following treatment with anti-IFN-γ antibodies or following IFNGR1 or STAT1 silencing. CONCLUSION IFN-γ contributes to the pathogenesis of SLE through the IFNGR1/2-pSTAT1-TBX21 axis and regulates inflammation and immune complex formation in SLE mice.
Collapse
Affiliation(s)
- Ying Chen
- Department of Nephrology, First Affiliated Hospital of China Medical UniversityShenyang 100012, Liaoning, China
| | - Bailing Tian
- Department of Rheumatology and Immunology, First Affiliated Hospital of China Medical UniversityShenyang 100012, Liaoning, China
| |
Collapse
|
21
|
Martínez-Gil N, Maneu V, Kutsyr O, Fernández-Sánchez L, Sánchez-Sáez X, Sánchez-Castillo C, Campello L, Lax P, Pinilla I, Cuenca N. Cellular and molecular alterations in neurons and glial cells in inherited retinal degeneration. Front Neuroanat 2022; 16:984052. [PMID: 36225228 PMCID: PMC9548552 DOI: 10.3389/fnana.2022.984052] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Accepted: 08/29/2022] [Indexed: 11/19/2022] Open
Abstract
Multiple gene mutations have been associated with inherited retinal dystrophies (IRDs). Despite the spectrum of phenotypes caused by the distinct mutations, IRDs display common physiopathology features. Cell death is accompanied by inflammation and oxidative stress. The vertebrate retina has several attributes that make this tissue vulnerable to oxidative and nitrosative imbalance. The high energy demands and active metabolism in retinal cells, as well as their continuous exposure to high oxygen levels and light-induced stress, reveal the importance of tightly regulated homeostatic processes to maintain retinal function, which are compromised in pathological conditions. In addition, the subsequent microglial activation and gliosis, which triggers the secretion of pro-inflammatory cytokines, chemokines, trophic factors, and other molecules, further worsen the degenerative process. As the disease evolves, retinal cells change their morphology and function. In disease stages where photoreceptors are lost, the remaining neurons of the retina to preserve their function seek out for new synaptic partners, which leads to a cascade of morphological alterations in retinal cells that results in a complete remodeling of the tissue. In this review, we describe important molecular and morphological changes in retinal cells that occur in response to oxidative stress and the inflammatory processes underlying IRDs.
Collapse
Affiliation(s)
- Natalia Martínez-Gil
- Department of Physiology, Genetics and Microbiology, University of Alicante, Alicante, Spain
| | - Victoria Maneu
- Department of Optics, Pharmacology and Anatomy, University of Alicante, Alicante, Spain
| | - Oksana Kutsyr
- Department of Physiology, Genetics and Microbiology, University of Alicante, Alicante, Spain
| | | | - Xavier Sánchez-Sáez
- Department of Physiology, Genetics and Microbiology, University of Alicante, Alicante, Spain
| | - Carla Sánchez-Castillo
- Department of Physiology, Genetics and Microbiology, University of Alicante, Alicante, Spain
| | - Laura Campello
- Department of Physiology, Genetics and Microbiology, University of Alicante, Alicante, Spain
| | - Pedro Lax
- Department of Physiology, Genetics and Microbiology, University of Alicante, Alicante, Spain
- Alicante Institute for Health and Biomedical Research (ISABIAL), Alicante, Spain
| | - Isabel Pinilla
- Aragón Institute for Health Research (IIS Aragón), Zaragoza, Spain
- Department of Ophthalmology, Lozano Blesa University Hospital, Zaragoza, Spain
- Department of Surgery, University of Zaragoza, Zaragoza, Spain
- Isabel Pinilla,
| | - Nicolás Cuenca
- Department of Physiology, Genetics and Microbiology, University of Alicante, Alicante, Spain
- Alicante Institute for Health and Biomedical Research (ISABIAL), Alicante, Spain
- Institute Ramón Margalef, University of Alicante, Alicante, Spain
- *Correspondence: Nicolás Cuenca,
| |
Collapse
|
22
|
Sharma P, Ramachandran R. Retina regeneration: lessons from vertebrates. OXFORD OPEN NEUROSCIENCE 2022; 1:kvac012. [PMID: 38596712 PMCID: PMC10913848 DOI: 10.1093/oons/kvac012] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 05/24/2022] [Accepted: 06/25/2022] [Indexed: 04/11/2024]
Abstract
Unlike mammals, vertebrates such as fishes and frogs exhibit remarkable tissue regeneration including the central nervous system. Retina being part of the central nervous system has attracted the interest of several research groups to explore its regenerative ability in different vertebrate models including mice. Fishes and frogs completely restore the size, shape and tissue structure of an injured retina. Several studies have unraveled molecular mechanisms underlying retina regeneration. In teleosts, soon after injury, the Müller glial cells of the retina reprogram to form a proliferating population of Müller glia-derived progenitor cells capable of differentiating into various neural cell types and Müller glia. In amphibians, the transdifferentiation of retinal pigment epithelium and differentiation of ciliary marginal zone cells contribute to retina regeneration. In chicks and mice, supplementation with external growth factors or genetic modifications cause a partial regenerative response in the damaged retina. The initiation of retina regeneration is achieved through sequential orchestration of gene expression through controlled modulations in the genetic and epigenetic landscape of the progenitor cells. Several developmental biology pathways are turned on during the Müller glia reprogramming, retinal pigment epithelium transdifferentiation and ciliary marginal zone differentiation. Further, several tumorigenic pathways and gene expression events also contribute to the complete regeneration cascade of events. In this review, we address the various retinal injury paradigms and subsequent gene expression events governed in different vertebrate species. Further, we compared how vertebrates such as teleost fishes and amphibians can achieve excellent regenerative responses in the retina compared with their mammalian counterparts.
Collapse
Affiliation(s)
- Poonam Sharma
- Department of Biological Sciences, Indian Institute of Science Education and Research, Mohali, Knowledge City, SAS Nagar, Sector 81, Manauli PO, 140306 Mohali, Punjab, India
| | - Rajesh Ramachandran
- Department of Biological Sciences, Indian Institute of Science Education and Research, Mohali, Knowledge City, SAS Nagar, Sector 81, Manauli PO, 140306 Mohali, Punjab, India
| |
Collapse
|
23
|
Peña JS, Vazquez M. Harnessing the Neuroprotective Behaviors of Müller Glia for Retinal Repair. FRONT BIOSCI-LANDMRK 2022; 27:169. [PMID: 35748245 PMCID: PMC9639582 DOI: 10.31083/j.fbl2706169] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Revised: 04/18/2022] [Accepted: 05/05/2022] [Indexed: 11/29/2022]
Abstract
Progressive and irreversible vision loss in mature and aging adults creates a health and economic burden, worldwide. Despite the advancements of many contemporary therapies to restore vision, few approaches have considered the innate benefits of gliosis, the endogenous processes of retinal repair that precede vision loss. Retinal gliosis is fundamentally driven by Müller glia (MG) and is characterized by three primary cellular mechanisms: hypertrophy, proliferation, and migration. In early stages of gliosis, these processes have neuroprotective potential to halt the progression of disease and encourage synaptic activity among neurons. Later stages, however, can lead to glial scarring, which is a hallmark of disease progression and blindness. As a result, the neuroprotective abilities of MG have remained incompletely explored and poorly integrated into current treatment regimens. Bioengineering studies of the intrinsic behaviors of MG hold promise to exploit glial reparative ability, while repressing neuro-disruptive MG responses. In particular, recent in vitro systems have become primary models to analyze individual gliotic processes and provide a stepping stone for in vivo strategies. This review highlights recent studies of MG gliosis seeking to harness MG neuroprotective ability for regeneration using contemporary biotechnologies. We emphasize the importance of studying gliosis as a reparative mechanism, rather than disregarding it as an unfortunate clinical prognosis in diseased retina.
Collapse
Affiliation(s)
- Juan S. Peña
- Department of Biomedical Engineering, Rutgers, The State
University of New Jersey, Piscataway (08854), New Jersey, USA
| | - Maribel Vazquez
- Department of Biomedical Engineering, Rutgers, The State
University of New Jersey, Piscataway (08854), New Jersey, USA
| |
Collapse
|
24
|
Palazzo I, Todd LJ, Hoang TV, Reh TA, Blackshaw S, Fischer AJ. NFkB-signaling promotes glial reactivity and suppresses Müller glia-mediated neuron regeneration in the mammalian retina. Glia 2022; 70:1380-1401. [PMID: 35388544 DOI: 10.1002/glia.24181] [Citation(s) in RCA: 46] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Revised: 03/28/2022] [Accepted: 03/29/2022] [Indexed: 12/25/2022]
Abstract
Müller glia (MG) in mammalian retinas are incapable of regenerating neurons after damage, whereas the MG in lower vertebrates regenerate functional neurons. Identification of cell signaling pathways and gene regulatory networks that regulate MG-mediated regeneration is key to harnessing the regenerative potential of MG. Here, we study how NFkB-signaling influences glial responses to damage and reprogramming of MG into neurons in the rodent retina. We find activation of NFkB and dynamic expression of NFkB-associated genes in MG after damage, however damage-induced NFkB activation is inhibited by microglia ablation. Knockout of NFkB in MG suppressed the accumulation of immune cells after damage. Inhibition of NFkB following NMDA-damage significantly enhanced the reprogramming of Ascl1-overexpressing MG into neuron-like cells. scRNA-seq of retinal glia following inhibition of NFkB reveals coordination with signaling via TGFβ2 and suppression of NFI and Id transcription factors. Inhibition of Smad3 signal transducer or Id transcription factors increased numbers of neuron-like cells produced by Ascl1-overexpressing MG. We conclude that NFkB is a key signaling hub that is activated in MG after damage, mediates the accumulation of immune cells, and suppresses the neurogenic potential of MG.
Collapse
Affiliation(s)
- Isabella Palazzo
- Department of Neuroscience, College of Medicine, The Ohio State University, Columbus, Ohio, USA
| | - Levi J Todd
- Department of Biological Structure, College of Medicine, University of Washington, Seattle, Washington, USA
| | - Thanh V Hoang
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Thomas A Reh
- Department of Biological Structure, College of Medicine, University of Washington, Seattle, Washington, USA
| | - Seth Blackshaw
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Andy J Fischer
- Department of Neuroscience, College of Medicine, The Ohio State University, Columbus, Ohio, USA
| |
Collapse
|