1
|
Badla O, Badla BA, Almobayed A, Mendoza C, Kishor K, Bhattacharya SK. Ischemic Optic Neuropathy: A Review of Current and Potential Future Pharmacotherapies. Pharmaceuticals (Basel) 2024; 17:1281. [PMID: 39458922 PMCID: PMC11510045 DOI: 10.3390/ph17101281] [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: 08/05/2024] [Revised: 09/13/2024] [Accepted: 09/25/2024] [Indexed: 10/28/2024] Open
Abstract
The treatment of arteritic anterior ischemic optic neuropathy (AAION), non-arteritic ischemic optic neuropathy (NAAION), and posterior ischemic optic neuropathy (PION) is a topic of ongoing research with mixed evidence on some pharmacotherapies and a need for more consensus. This manuscript provides an overview of these conditions' current, potential future, and attempted pharmacotherapies. AAION's current treatment regimen consists of high-dose steroids, with methotrexate, tocilizumab, and abatacept, being the most viable steroid-sparing therapy candidates. As for NAAION, the treatments being tried are vast, with mixed evidence supporting each modality. Similarly, despite the various treatment options explored, there still needs to be a universally effective therapy for PION. More research is needed to formulate an agreed-upon treatment regimen for these conditions.
Collapse
Affiliation(s)
- Omar Badla
- Department of Ophthalmology, Bascom Palmer Eye Institute, University of Miami Miller School Medicine, Miami, FL 33136, USA (A.A.)
- Miami Integrative Metabolomics Research Center, Miami, FL 33136, USA
| | - Beshr Abdulaziz Badla
- College of Medicine, Mohammed Bin Rashid University of Medicine and Health Sciences, Dubai P.O. Box 505055, United Arab Emirates;
| | - Amr Almobayed
- Department of Ophthalmology, Bascom Palmer Eye Institute, University of Miami Miller School Medicine, Miami, FL 33136, USA (A.A.)
- Miami Integrative Metabolomics Research Center, Miami, FL 33136, USA
| | - Carlos Mendoza
- Department of Ophthalmology, Bascom Palmer Eye Institute, University of Miami Miller School Medicine, Miami, FL 33136, USA (A.A.)
| | - Krishna Kishor
- Department of Ophthalmology, Bascom Palmer Eye Institute, University of Miami Miller School Medicine, Miami, FL 33136, USA (A.A.)
- Miami Integrative Metabolomics Research Center, Miami, FL 33136, USA
| | - Sanjoy K. Bhattacharya
- Department of Ophthalmology, Bascom Palmer Eye Institute, University of Miami Miller School Medicine, Miami, FL 33136, USA (A.A.)
- Miami Integrative Metabolomics Research Center, Miami, FL 33136, USA
- Graduate Program in Molecular and Cellular Pharmacology, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| |
Collapse
|
2
|
Singh M, Krishnamoorthy VR, Kim S, Khurana S, LaPorte HM. Brain-derived neuerotrophic factor and related mechanisms that mediate and influence progesterone-induced neuroprotection. Front Endocrinol (Lausanne) 2024; 15:1286066. [PMID: 38469139 PMCID: PMC10925611 DOI: 10.3389/fendo.2024.1286066] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Accepted: 01/15/2024] [Indexed: 03/13/2024] Open
Abstract
Historically, progesterone has been studied significantly within the context of reproductive biology. However, there is now an abundance of evidence for its role in regions of the central nervous system (CNS) associated with such non-reproductive functions that include cognition and affect. Here, we describe mechanisms of progesterone action that support its brain-protective effects, and focus particularly on the role of neurotrophins (such as brain-derived neurotrophic factor, BDNF), the receptors that are critical for their regulation, and the role of certain microRNA in influencing the brain-protective effects of progesterone. In addition, we describe evidence to support the particular importance of glia in mediating the neuroprotective effects of progesterone. Through this review of these mechanisms and our own prior published work, we offer insight into why the effects of a progestin on brain protection may be dependent on the type of progestin (e.g., progesterone versus the synthetic, medroxyprogesterone acetate) used, and age, and as such, we offer insight into the future clinical implication of progesterone treatment for such disorders that include Alzheimer's disease, stroke, and traumatic brain injury.
Collapse
Affiliation(s)
- Meharvan Singh
- Department of Cell and Molecular Physiology, Stritch School of Medicine, Loyola University Chicago, Maywood, IL, United States
| | | | | | | | | |
Collapse
|
3
|
Budihardja BM, Anggraini E, Pratiwi RW, Nastiti AD, Nusanti S. Neuroprotective Strategies for Nonarteritic Anterior Ischemic Optic Neuropathy: A Systematic Review. KOREAN JOURNAL OF OPHTHALMOLOGY 2023; 37:328-339. [PMID: 37563973 PMCID: PMC10427903 DOI: 10.3341/kjo.2022.0166] [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: 12/28/2022] [Revised: 06/08/2023] [Accepted: 07/22/2023] [Indexed: 08/12/2023] Open
Abstract
PURPOSE Nonarteritic anterior ischemic optic neuropathy (NAION) is the second most common form of optic neuropathy. Most patients show no improvement over time. Until now, there is still no definitive therapy for NAION. The available literatures on the possible treatment of NAION are quite diverse and controversial. Neuroprotection strategies have been suggested as one of the potential treatments for NAION. This review aims to critically evaluate the literature on neuroprotective strategy for NAION. METHODS This report was written in accordance with PRISMA (Preferred Reporting Items for Systematic Reviews and Meta-Analysis) guidelines. We performed a systematic literature search in Pubmed, Science Direct, Proquest, and Cochrane databases. Only neuroprotective agents that directly work in protecting neurons were included. The outcome of interest in this review is retinal ganglion cell density and apoptosis for animal studies and retinal nerve fiber layer thickness for human studies. RESULTS The systematic search identified 591 studies of which 24 met the eligibility criteria, including 21 animal studies and three human studies. Only a few of the studies evaluated the same treatments, showing how diverse neuroprotector treatments are currently being evaluated as NAION treatment. From 21 animal studies, 14 studies showed significantly higher retinal ganglion cell density (1.49- to 2.81-fold) with neuroprotective treatment compared to control group. Two of three human studies in this review had also found a beneficial effect of preserving retinal nerve fiber layer thickness in NAION patients. CONCLUSIONS This review suggests the potential of neuroprotection as a viable option in the quest for an effective treatment strategy for NAION. Further studies, particularly clinical studies, are necessary to establish its efficacy in NAION patients.
Collapse
Affiliation(s)
- Brigitta Marcia Budihardja
- Department of Ophthalmology, Dr. Cipto Mangunkusumo Hospital, Faculty of Medicine, University of Indonesia, Jakarta,
Indonesia
| | - Erika Anggraini
- Department of Ophthalmology, Dr. Cipto Mangunkusumo Hospital, Faculty of Medicine, University of Indonesia, Jakarta,
Indonesia
| | - Rianti Wulandari Pratiwi
- Department of Ophthalmology, Dr. Cipto Mangunkusumo Hospital, Faculty of Medicine, University of Indonesia, Jakarta,
Indonesia
| | - Anya Dewi Nastiti
- Department of Ophthalmology, Dr. Cipto Mangunkusumo Hospital, Faculty of Medicine, University of Indonesia, Jakarta,
Indonesia
| | - Syntia Nusanti
- Division of Neuro-Ophthalmology, Department of Ophthalmology, Dr. Cipto Mangunkusumo Hospital, Faculty of Medicine, University of Indonesia, Jakarta,
Indonesia
| |
Collapse
|
4
|
Allen RS, Khayat CT, Feola AJ, Win AS, Grubman AR, Chesler KC, He L, Dixon JA, Kern TS, Iuvone PM, Thule PM, Pardue MT. Diabetic rats with high levels of endogenous dopamine do not show retinal vascular pathology. Front Neurosci 2023; 17:1125784. [PMID: 37034167 PMCID: PMC10073440 DOI: 10.3389/fnins.2023.1125784] [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: 12/16/2022] [Accepted: 02/21/2023] [Indexed: 04/11/2023] Open
Abstract
Purpose Limited research exists on the time course of long-term retinal and cerebral deficits in diabetic rodents. Previously, we examined short term (4-8 weeks) deficits in the Goto-Kakizaki (GK) rat model of Type II diabetes. Here, we investigated the long-term (1-8 months) temporal appearance of functional deficits (retinal, cognitive, and motor), retinal vascular pathology, and retinal dopamine levels in the GK rat. Methods In GK rats and Wistar controls, retinal neuronal function (electroretinogram), cognitive function (Y-maze), and motor function (rotarod) were measured at 1, 2, 4, 6, and 8 months of age. In addition, we evaluated retinal vascular function (functional hyperemia) and glucose and insulin tolerance. Retinas from rats euthanized at ≥8 months were assessed for vascular pathology. Dopamine and DOPAC levels were measured via HPLC in retinas from rats euthanized at 1, 2, 8, and 12 months. Results Goto-Kakizaki rats exhibited significant glucose intolerance beginning at 4 weeks and worsening over time (p < 0.001). GK rats also showed significant delays in flicker and oscillatory potential implicit times (p < 0.05 to p < 0.001) beginning at 1 month. Cognitive deficits were observed beginning at 6 months (p < 0.05), but no motor deficits. GK rats showed no deficits in functional hyperemia and no increase in acellular retinal capillaries. Dopamine levels were twice as high in GK vs. Wistar retinas at 1, 2, 8, and 12 months (p < 0.001). Conclusion As shown previously, retinal deficits were detectable prior to cognitive deficits in GK rats. While retinal neuronal function was compromised, retinal vascular pathology was not observed, even at 12+ months. High endogenous levels of dopamine in the GK rat may be acting as an anti-angiogenic and providing protection against vascular pathology.
Collapse
Affiliation(s)
- Rachael S. Allen
- Atlanta VA Center for Visual and Neurocognitive Rehabilitation, Atlanta VA Healthcare System, Decatur, GA, United States
- Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, United States
| | - Cara T. Khayat
- Atlanta VA Center for Visual and Neurocognitive Rehabilitation, Atlanta VA Healthcare System, Decatur, GA, United States
| | - Andrew J. Feola
- Atlanta VA Center for Visual and Neurocognitive Rehabilitation, Atlanta VA Healthcare System, Decatur, GA, United States
- Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, United States
- Department of Ophthalmology, Emory University, Atlanta, GA, United States
| | - Alice S. Win
- Atlanta VA Center for Visual and Neurocognitive Rehabilitation, Atlanta VA Healthcare System, Decatur, GA, United States
- Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, United States
| | - Allison R. Grubman
- Atlanta VA Center for Visual and Neurocognitive Rehabilitation, Atlanta VA Healthcare System, Decatur, GA, United States
- Department of Ophthalmology, Emory University, Atlanta, GA, United States
| | - Kyle C. Chesler
- Atlanta VA Center for Visual and Neurocognitive Rehabilitation, Atlanta VA Healthcare System, Decatur, GA, United States
- Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, United States
| | - Li He
- Department of Ophthalmology, Emory University, Atlanta, GA, United States
- Department of Pharmacology and Chemical Biology, Emory University, Atlanta, GA, United States
| | - Jendayi A. Dixon
- Department of Ophthalmology, Emory University, Atlanta, GA, United States
| | - Timothy S. Kern
- Department of Pharmacology, Case Western Reserve University, Cleveland, OH, United States
- Veterans Administration Medical Center Research Service, Cleveland, OH, United States
- Gavin Herbert Eye Institute, University of California, Irvine, Irvine, CA, United States
| | - P. Michael Iuvone
- Department of Ophthalmology, Emory University, Atlanta, GA, United States
- Department of Pharmacology and Chemical Biology, Emory University, Atlanta, GA, United States
| | - Peter M. Thule
- Section Endocrinology and Metabolism, Atlanta VA Medical Center, Emory University School of Medicine, Decatur, GA, United States
| | - Machelle T. Pardue
- Atlanta VA Center for Visual and Neurocognitive Rehabilitation, Atlanta VA Healthcare System, Decatur, GA, United States
- Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, United States
- Department of Ophthalmology, Emory University, Atlanta, GA, United States
| |
Collapse
|
5
|
Douglass A, Dattilo M, Feola AJ. Evidence for Menopause as a Sex-Specific Risk Factor for Glaucoma. Cell Mol Neurobiol 2023; 43:79-97. [PMID: 34981287 PMCID: PMC9250947 DOI: 10.1007/s10571-021-01179-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Accepted: 12/03/2021] [Indexed: 01/07/2023]
Abstract
Glaucoma is a leading cause of irreversible blindness worldwide and is characterized by progressive loss of visual function and retinal ganglion cells (RGC). Current epidemiological, clinical, and basic science evidence suggest that estrogen plays a role in the aging of the optic nerve. Menopause, a major biological life event affecting all women, coincides with a decrease in circulating sex hormones, such as estrogen. While 59% of the glaucomatous population are females, sex is not considered a risk factor for developing glaucoma. In this review, we explore whether menopause is a sex-specific risk factor for glaucoma. First, we investigate how menopause is defined as a sex-specific risk factor for other pathologies, including cardiovascular disease, osteoarthritis, and bone health. Next, we discuss clinical evidence that highlights the potential role of menopause in glaucoma. We also highlight preclinical studies that demonstrate larger vision and RGC loss following surgical menopause and how estrogen is protective in models of RGC injury. Lastly, we explore how surgical menopause and estrogen signaling are related to risk factors associated with developing glaucoma (e.g., intraocular pressure, aqueous outflow resistance, and ocular biomechanics). We hypothesize that menopause potentially sets the stage to develop glaucoma and therefore is a sex-specific risk factor for this disease.
Collapse
Affiliation(s)
- Amber Douglass
- grid.484294.7Center for Visual and Neurocognitive Rehabilitation, Atlanta VA Healthcare System, Decatur, GA USA
| | - Michael Dattilo
- grid.189967.80000 0001 0941 6502Department of Ophthalmology, Emory Eye Center, Emory University School of Medicine, B2503, Clinic B Building, 1365B Clifton Road NE, Atlanta, GA 30322 USA ,grid.414026.50000 0004 0419 4084Department of Ophthalmology, Atlanta Veterans Affairs Medical Center, Atlanta, GA USA ,grid.213917.f0000 0001 2097 4943Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA USA
| | - Andrew J. Feola
- grid.484294.7Center for Visual and Neurocognitive Rehabilitation, Atlanta VA Healthcare System, Decatur, GA USA ,grid.189967.80000 0001 0941 6502Department of Ophthalmology, Emory Eye Center, Emory University School of Medicine, B2503, Clinic B Building, 1365B Clifton Road NE, Atlanta, GA 30322 USA ,grid.213917.f0000 0001 2097 4943Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA USA
| |
Collapse
|
6
|
Jeon J, Kim SH, Kong E, Kim SJ, Yang JM, Lee JY, Lee J, Kim YM, Kim P. Establishment of the reproducible branch retinal artery occlusion mouse model and intravital longitudinal imaging of the retinal CX3CR1-GFP+ cells after spontaneous arterial recanalization. Front Med (Lausanne) 2022; 9:897800. [PMID: 35911406 PMCID: PMC9334526 DOI: 10.3389/fmed.2022.897800] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Accepted: 06/29/2022] [Indexed: 11/23/2022] Open
Abstract
Animal models of retinal artery occlusion (RAO) have been widely used in many studies. However, most of these studies prefer using a central retinal artery occlusion (CRAO) which is a typical global ischemia model of the retina, due to the technical limitation of producing single vessel targeted modeling with real-time imaging. A focal ischemia model, such as branch retinal artery occlusion (BRAO), is also needed for explaining interactions, including the immunological reaction between the ischemic retina and adjacent healthy retina. Accordingly, a relevant model for clinical RAO patients has been demanded to understand the pathophysiology of the RAO disease. Herein, we establish a convenient BRAO mouse model to research the focal reaction of the retina. As a photo-thrombotic agent, Rose bengal was intravenously injected into 7 week-old transgenic mice (CX3CR1-GFP) for making embolism occlusion, which causes pathology similarly to clinical cases. In an optimized condition, a 561 nm laser (13.1 mw) was projected to a targeted vessel to induce photo-thrombosis for 27 s by custom-built retinal confocal microscopy. Compared to previous BRAO models, the procedures of thrombosis generation were naturally and minimal invasively generated with real-time retinal imaging. In addition, by utilizing the self-remission characteristics of Rose bengal thrombus, a reflow of the BRAO with immunological reactions of the CX3CR1-GFP+ inflammatory cells such as the retinal microglia and monocytes was monitored and analyzed. In this models, reperfusion began on day 3 after modeling. Simultaneously, the activation of CX3CR1-GFP+ inflammatory cells, including the increase of activation marker and morphologic change, was confirmed by immunohistochemical (IHC) staining and quantitative real-time PCR. CD86 and Nox2 were prominently expressed on day 3 after the modeling. At day 7, blood flow was almost restored in the large vessels. CX3CR1-GFP+ populations in both superficial and deep layers of the retina also increased around even in the BRAO peri-ischemic area. In summary, this study successfully establishes a reproducible BRAO modeling method with convenient capabilities of easily controllable time points and selection of a specific single vessel. It can be a useful tool to analyze the behavior of inflammatory cell after spontaneous arterial recanalization in BRAO and further investigate the pathophysiology of BRAO.
Collapse
Affiliation(s)
- Jehwi Jeon
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, South Korea
- KI for Health Science and Technology (KIHST), Korea Advanced Institute of Science and Technology (KAIST), Daejeon, South Korea
| | - Sang-Hoon Kim
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, South Korea
| | - Eunji Kong
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, South Korea
- KI for Health Science and Technology (KIHST), Korea Advanced Institute of Science and Technology (KAIST), Daejeon, South Korea
| | - Soo Jin Kim
- Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea
| | - Jee Myung Yang
- Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea
- Dongguk University Ilsan Hospital, Ilsan, South Korea
| | - Joo Yong Lee
- Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea
| | - Junyeop Lee
- Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea
| | - You-Me Kim
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, South Korea
| | - Pilhan Kim
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, South Korea
- KI for Health Science and Technology (KIHST), Korea Advanced Institute of Science and Technology (KAIST), Daejeon, South Korea
- *Correspondence: Pilhan Kim,
| |
Collapse
|
7
|
Pinilla I, Maneu V, Campello L, Fernández-Sánchez L, Martínez-Gil N, Kutsyr O, Sánchez-Sáez X, Sánchez-Castillo C, Lax P, Cuenca N. Inherited Retinal Dystrophies: Role of Oxidative Stress and Inflammation in Their Physiopathology and Therapeutic Implications. Antioxidants (Basel) 2022; 11:antiox11061086. [PMID: 35739983 PMCID: PMC9219848 DOI: 10.3390/antiox11061086] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2022] [Revised: 05/24/2022] [Accepted: 05/26/2022] [Indexed: 12/13/2022] Open
Abstract
Inherited retinal dystrophies (IRDs) are a large group of genetically and clinically heterogeneous diseases characterized by the progressive degeneration of the retina, ultimately leading to loss of visual function. Oxidative stress and inflammation play fundamental roles in the physiopathology of these diseases. Photoreceptor cell death induces an inflammatory state in the retina. The activation of several molecular pathways triggers different cellular responses to injury, including the activation of microglia to eliminate debris and recruit inflammatory cells from circulation. Therapeutical options for IRDs are currently limited, although a small number of patients have been successfully treated by gene therapy. Many other therapeutic strategies are being pursued to mitigate the deleterious effects of IRDs associated with oxidative metabolism and/or inflammation, including inhibiting reactive oxygen species’ accumulation and inflammatory responses, and blocking autophagy. Several compounds are being tested in clinical trials, generating great expectations for their implementation. The present review discusses the main death mechanisms that occur in IRDs and the latest therapies that are under investigation.
Collapse
Affiliation(s)
- Isabel Pinilla
- Aragón Health Research Institute (IIS Aragón), 50009 Zaragoza, Spain
- Department of Ophthalmology, Lozano Blesa, University Hospital, 50009 Zaragoza, Spain
- Department of Surgery, University of Zaragoza, 50009 Zaragoza, Spain
- Correspondence: (I.P.); (V.M.)
| | - Victoria Maneu
- Department of Optics, Pharmacology and Anatomy, University of Alicante, 03690 Alicante, Spain;
- Alicante Institute for Health and Biomedical Research (ISABIAL), 03010 Alicante, Spain; (P.L.); (N.C.)
- Correspondence: (I.P.); (V.M.)
| | - Laura Campello
- Department of Physiology, Genetics and Microbiology, University of Alicante, 03690 Alicante, Spain; (L.C.); (N.M.-G.); (O.K.); (X.S.-S.); (C.S.-C.)
| | - Laura Fernández-Sánchez
- Department of Optics, Pharmacology and Anatomy, University of Alicante, 03690 Alicante, Spain;
| | - Natalia Martínez-Gil
- Department of Physiology, Genetics and Microbiology, University of Alicante, 03690 Alicante, Spain; (L.C.); (N.M.-G.); (O.K.); (X.S.-S.); (C.S.-C.)
| | - Oksana Kutsyr
- Department of Physiology, Genetics and Microbiology, University of Alicante, 03690 Alicante, Spain; (L.C.); (N.M.-G.); (O.K.); (X.S.-S.); (C.S.-C.)
| | - Xavier Sánchez-Sáez
- Department of Physiology, Genetics and Microbiology, University of Alicante, 03690 Alicante, Spain; (L.C.); (N.M.-G.); (O.K.); (X.S.-S.); (C.S.-C.)
| | - Carla Sánchez-Castillo
- Department of Physiology, Genetics and Microbiology, University of Alicante, 03690 Alicante, Spain; (L.C.); (N.M.-G.); (O.K.); (X.S.-S.); (C.S.-C.)
| | - Pedro Lax
- Alicante Institute for Health and Biomedical Research (ISABIAL), 03010 Alicante, Spain; (P.L.); (N.C.)
- Department of Physiology, Genetics and Microbiology, University of Alicante, 03690 Alicante, Spain; (L.C.); (N.M.-G.); (O.K.); (X.S.-S.); (C.S.-C.)
| | - Nicolás Cuenca
- Alicante Institute for Health and Biomedical Research (ISABIAL), 03010 Alicante, Spain; (P.L.); (N.C.)
- Department of Physiology, Genetics and Microbiology, University of Alicante, 03690 Alicante, Spain; (L.C.); (N.M.-G.); (O.K.); (X.S.-S.); (C.S.-C.)
| |
Collapse
|
8
|
Yang P, Chen L, Shi Y, Zhou F, Tian H, Li J, Gao L. Progesterone alters the activation and typing of the microglia in the optic nerve crush model. Exp Eye Res 2021; 212:108805. [PMID: 34699875 DOI: 10.1016/j.exer.2021.108805] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Revised: 10/18/2021] [Accepted: 10/21/2021] [Indexed: 12/21/2022]
Abstract
Microglia have a protective effect on the central nervous system (CNS), but their over-proliferation can cause secondary injury to the retina following optic nerve crush (ONC). Progesterone as a steroid gonadal hormone has been used in some experimental animal models for its neuroprotective effect. However, there is limited attention on the interactions between progesterone and microglia in retinal diseases. This study investigated the proliferation, morphology changes, and cell types of microglia at 3 days and 7 days after ONC. We found that progesterone treatment in unilateral optic nerve injury mice significantly reduced densities and morphological change of microglia at 7 days in the ganglion cell layer (GCL), especially in the retinal central. Inhibition of the microglia proliferation and transformation of ramified microglia into ameboid macrophages also appeared in the inner plexiform layer (IPL). Moreover, progesterone also regulated the TNF signal pathway, which was similar to the specific elimination of the M1 phenotype. M1 marks such as tumor necrosis factor alpha (TNF-α), inducible NOS(iNOS), interleukin-6 (IL-6), and Fc receptor (CD16 and CD32) significantly downregulated by progesterone treatment whether at 3 days or 7 days after ONC. On the other hand, progesterone continuously increased the expression of the M2 marks, including interleukin-4 (IL-4), arginase 1 (Arg1), and mannose receptor (CD206) since the third day, while the expression levels of transforming growth factor (TGF-β) only increased at 7 days. In general, this study elucidated the mechanism that progesterone prevented further damage on the retina by inhibiting proliferation, activation, and changing the type of microglia.
Collapse
Affiliation(s)
- Pengfei Yang
- School of Life Sciences, Lanzhou University, Lanzhou, 730000, China
| | - Linchi Chen
- School of Life Sciences, Lanzhou University, Lanzhou, 730000, China
| | - Yongpeng Shi
- School of Life Sciences, Lanzhou University, Lanzhou, 730000, China
| | - Fangfang Zhou
- College of Pastoral Agriculture Science and Technology, Lanzhou University, Lanzhou, 730020, China
| | - Huanbing Tian
- School of Life Sciences, Lanzhou University, Lanzhou, 730000, China
| | - Jiande Li
- School of Life Sciences, Lanzhou University, Lanzhou, 730000, China
| | - Lan Gao
- School of Life Sciences, Lanzhou University, Lanzhou, 730000, China.
| |
Collapse
|
9
|
Sun X, Hu Y, Zhou H, Wang S, Zhou C, Lin L, Zhu T, Ge J, Han J, Zhou Y, Jin G, Wang Y, Zu J, Shi H, Yang X, Zan K, Wang J, Hua F. Inhibition of progesterone receptor membrane component-1 exacerbates neonatal hypoxic-ischemic cerebral damage in male mice. Exp Neurol 2021; 347:113893. [PMID: 34653511 DOI: 10.1016/j.expneurol.2021.113893] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2021] [Revised: 10/04/2021] [Accepted: 10/08/2021] [Indexed: 11/04/2022]
Abstract
This study investigated the expression of progesterone receptor membrane component 1 (pgrmc1) in the brains of male and female mice, and the effect of inhibiting pgrmc1 on neonatal hypoxic-ischemic (HI) cerebral injury in male mice. A mouse model of neonatal HI brain injury was established, and AG205, a specific antagonist of pgrmc1, was injected into the left lateral cerebral ventricle 1 h before HI. Histological staining, behavior testing, Western blots, and quantitative PCR (qPCR) were employed to evaluate pgrmc1 expression, brain damage, neurological function, and molecular mechanisms. Results demonstrated that the mRNA and protein levels of pgrmc1 increased significantly in the cortex and hippocampus 72 h after HI without sex differences. The inhibition of pgrmc1 exacerbated the neonatal brain damage in the acute stage of HI in male mice as seen in the increase in brain water content, infarction area, and neuronal death. Inhibition of pgrmc1 also aggravated the neurological dysfunction and anxiety induced by HI brain injury. In addition, inhibition of pgrmc1 activated the NF-kB signaling and NF-κB-mediated cytokines, and inhibited BDNF/PI3K/AKT pathway in the brains of the newborn HI mice. The results indicated that pgrmc1 inhibition exacerbated the brain damage in newborn male mice subjected to HI by activating IκBα/NFκB signaling and inhibiting BDNF/PI3K/Akt pathway.
Collapse
Affiliation(s)
- Xiaoyu Sun
- Department of Neurology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, China; Department of Rehabilitation Medicine, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Yuting Hu
- Department of Neurology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, China; Department of Rehabilitation Medicine, Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Hui Zhou
- Department of Neurology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, China; Institute of Neurological Diseases, Xuzhou Medical University, Xuzhou, China
| | - Shang Wang
- Department of Neurology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, China; Institute of Neurological Diseases, Xuzhou Medical University, Xuzhou, China
| | - Chao Zhou
- Department of Neurology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, China; Institute of Neurological Diseases, Xuzhou Medical University, Xuzhou, China
| | - Li Lin
- Department of Neurology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, China; Institute of Neurological Diseases, Xuzhou Medical University, Xuzhou, China
| | - Taiyang Zhu
- Department of Neurology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, China; Institute of Neurological Diseases, Xuzhou Medical University, Xuzhou, China
| | - Ji Ge
- Department of Neurology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, China; Institute of Neurological Diseases, Xuzhou Medical University, Xuzhou, China
| | - Jingjing Han
- Department of Neurology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, China; Institute of Neurological Diseases, Xuzhou Medical University, Xuzhou, China
| | - Yan Zhou
- Department of Neurology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, China; Institute of Neurological Diseases, Xuzhou Medical University, Xuzhou, China
| | - Guoliang Jin
- Department of Neurology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, China; Institute of Neurological Diseases, Xuzhou Medical University, Xuzhou, China
| | - Yuqiao Wang
- Department of Neurology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, China; Institute of Neurological Diseases, Xuzhou Medical University, Xuzhou, China
| | - Jie Zu
- Department of Neurology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, China; Institute of Neurological Diseases, Xuzhou Medical University, Xuzhou, China
| | - Hongjuan Shi
- Department of Neurology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, China; Institute of Neurological Diseases, Xuzhou Medical University, Xuzhou, China
| | - Xingxing Yang
- Department of Neurology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, China; Institute of Neurological Diseases, Xuzhou Medical University, Xuzhou, China
| | - Kun Zan
- Department of Neurology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, China; Institute of Neurological Diseases, Xuzhou Medical University, Xuzhou, China
| | - Jun Wang
- Department of Pediatrics, Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Fang Hua
- Department of Neurology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, China; Institute of Neurological Diseases, Xuzhou Medical University, Xuzhou, China.
| |
Collapse
|
10
|
Enz TJ, Tribble JR, Williams PA. Comparison of Glaucoma-Relevant Transcriptomic Datasets Identifies Novel Drug Targets for Retinal Ganglion Cell Neuroprotection. J Clin Med 2021; 10:3938. [PMID: 34501387 PMCID: PMC8432026 DOI: 10.3390/jcm10173938] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Revised: 08/10/2021] [Accepted: 08/27/2021] [Indexed: 12/20/2022] Open
Abstract
Glaucoma is a leading cause of blindness and is characterized by the progressive dysfunction and irreversible death of retinal ganglion cells. We aimed to identify shared differentially expressed genes (DE genes) between different glaucoma relevant models of retinal ganglion cell injury using existing RNA-sequencing data, thereby discovering targets for neuroprotective therapies. A comparison of DE genes from publicly available transcriptomic datasets identified 12 shared DE genes. The Comparative Toxicogenomics Database (CTD) was screened for compounds targeting a significant proportion of the identified DE genes. Forty compounds were identified in the CTD that interact with >50% of these shared DE genes. We next validated this approach by testing select compounds for an effect on retinal ganglion cell survival using a mouse retinal explant model. Folic acid, genistein, SB-431542, valproic acid, and WY-14643 (pirinixic acid) were tested. Folic acid, valproic acid, and WY-14643 demonstrated significant protection against retinal ganglion cell death in this model. The increasing prevalence of open access-omics data presents a resource to discover targets for future therapeutic investigation.
Collapse
Affiliation(s)
| | | | - Pete A. Williams
- Department of Clinical Neuroscience, Division of Eye and Vision, St. Erik Eye Hospital, Karolinska Institutet, 171 64 Stockholm, Sweden; (T.J.E.); (J.R.T.)
| |
Collapse
|
11
|
Alambiaga-Caravaca AM, Domenech-Monsell IM, Sebastián-Morelló M, Calatayud-Pascual MA, Merino V, Rodilla V, López-Castellano A. Development, characterization, and ex vivo evaluation of an insert for the ocular administration of progesterone. Int J Pharm 2021; 606:120921. [PMID: 34303817 DOI: 10.1016/j.ijpharm.2021.120921] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Revised: 07/06/2021] [Accepted: 07/19/2021] [Indexed: 01/05/2023]
Abstract
Progesterone (PG) affords neuroprotection in degenerative diseases associated to oxidative stress, such as cataracts, age-related macular degeneration, glaucoma, diabetic retinopathy and retinitis pigmentosa. The aim of this project was to develop ocular inserts for delivery of PG to the eye. Different inserts with PG in its composition were formulated and the insert with the best characteristics (59% polyvinyl alcohol, 39% polyvinylpyrrolidone K30 and 2% propylene glycol) was selected for ex vivo studies. Physical characteristics and drug release patterns of the insert were analysed. In vitro diffusion studies revealed a controlled diffusion of progesterone. Ex vivo experiments demonstrated similar trans-corneal and trans-scleral PG diffusion (corneal apparent permeability coefficient 6.46 ± 0.38 × 10-7 cm/s and scleral apparent permeability coefficient 5.87 ± 1.18 × 10-7 cm/s; mean ± SD; n = 5). However, the amount of PG accumulated in scleras was statistically higher than in corneas (30.07 ± 9.09 μg/cm2 and 15.56 ± 4.36 μg/cm2 respectively). The PG-loaded inserts (55.6 μg/cm2) were thin, translucent, showed no irritancy (HET-CAM test) and were elastic and robust, all suitable properties for its potential use in the treatment of several ocular diseases.
Collapse
Affiliation(s)
- Adrián M Alambiaga-Caravaca
- Department of Pharmacy, Faculty of Health Sciences, Institute of Biomedical sciences, Cardenal Herrera-CEU University, CEU Universities, C/Santiago Ramón y Cajal, s/n., Alfara del Patriarca 46115, Valencia, Spain
| | - Iris M Domenech-Monsell
- Department of Pharmacy, Faculty of Health Sciences, Institute of Biomedical sciences, Cardenal Herrera-CEU University, CEU Universities, C/Santiago Ramón y Cajal, s/n., Alfara del Patriarca 46115, Valencia, Spain
| | - María Sebastián-Morelló
- Department of Pharmacy, Faculty of Health Sciences, Institute of Biomedical sciences, Cardenal Herrera-CEU University, CEU Universities, C/Santiago Ramón y Cajal, s/n., Alfara del Patriarca 46115, Valencia, Spain
| | - M Aracely Calatayud-Pascual
- Department of Pharmacy, Faculty of Health Sciences, Institute of Biomedical sciences, Cardenal Herrera-CEU University, CEU Universities, C/Santiago Ramón y Cajal, s/n., Alfara del Patriarca 46115, Valencia, Spain
| | - Virginia Merino
- Department of Pharmacy and Pharmaceutical Technology and Parasitology, Faculty of Pharmacy. Institute of Molecular Recognition and Technological Development, Polytechnic University of Valencia, University of València, Valencia, Spain
| | - Vicent Rodilla
- Department of Pharmacy, Faculty of Health Sciences, Institute of Biomedical sciences, Cardenal Herrera-CEU University, CEU Universities, C/Santiago Ramón y Cajal, s/n., Alfara del Patriarca 46115, Valencia, Spain.
| | - Alicia López-Castellano
- Department of Pharmacy, Faculty of Health Sciences, Institute of Biomedical sciences, Cardenal Herrera-CEU University, CEU Universities, C/Santiago Ramón y Cajal, s/n., Alfara del Patriarca 46115, Valencia, Spain.
| |
Collapse
|
12
|
Sun S, Wang Y, Zhou Y, Ma W, Huang Y, Hu J, Wang Y. Serum progesterone and retinopathy in male patients with type 2 diabetes: A cross-sectional study. J Diabetes Investig 2021; 12:1228-1235. [PMID: 33202100 PMCID: PMC8264397 DOI: 10.1111/jdi.13463] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Revised: 11/11/2020] [Accepted: 11/12/2020] [Indexed: 11/28/2022] Open
Abstract
AIMS/INTRODUCTION The aim of this study was to investigate the relationship between serum progesterone (P) and retinopathy in male patients with type 2 diabetes mellitus, and to investigate whether P is associated with its progression. MATERIALS AND METHODS A total of 1,376 male participants with type 2 diabetes mellitus were recruited from Affiliated Hospital of Medical College Qingdao University (Qingdao, China). Through logistic regression analysis after adjusting the potential confounding variation, the odds ratio (OR) and the corresponding 95% confidence interval related to the quartiles of progesterone were obtained. RESULTS According to the quartiles of P levels, the prevalence rate of diabetic retinopathy (DR) in the last quartile is obviously greater to other quartiles (52.5-34.9%, 31.9%, 37.5%, P < 0.001). Compared with those in the first quartile, the prevalence of DR for the last quartile had an OR of 1.85 in the non-proliferative diabetic retinopathy group, while the OR was 8.35 in the proliferative diabetic retinopathy group (P < 0.001, unadjusted model). When adjusted for age, body mass index, duration of type 2 diabetes mellitus, glycated hemoglobin, blood pressure and other variables, the ORs for DR in the fourth quartile were 2.13 (95% confidence interval 1.49-3.06) in the non-proliferative diabetic retinopathy group and 8.44 (95% confidence interval 2.69-26.43) in the proliferative diabetic retinopathy group (P < 0.001). The positive association between P and DR risk was independent in adjusted logistic regression. CONCLUSIONS High levels of serum progesterone are significantly associated with DR in male hospitalized patients. This could mean that a higher P level in men is a potential clinical factor to identify DR, and the causality remains to be further explored.
Collapse
Affiliation(s)
- Shengnan Sun
- Department of EndocrinologyAffiliated Hospital of Medical College Qingdao UniversityQingdaoChina
| | - Yahao Wang
- Department of EndocrinologyAffiliated Hospital of Medical College Qingdao UniversityQingdaoChina
| | - Yue Zhou
- Department of EndocrinologyAffiliated Hospital of Medical College Qingdao UniversityQingdaoChina
| | - Wenru Ma
- Department of EndocrinologyAffiliated Hospital of Medical College Qingdao UniversityQingdaoChina
| | - Yajing Huang
- Department of EndocrinologyAffiliated Hospital of Medical College Qingdao UniversityQingdaoChina
| | - Jianxia Hu
- Department of EndocrinologyAffiliated Hospital of Medical College Qingdao UniversityQingdaoChina
| | - Yangang Wang
- Department of EndocrinologyAffiliated Hospital of Medical College Qingdao UniversityQingdaoChina
| |
Collapse
|
13
|
Alambiaga-Caravaca AM, Calatayud-Pascual MA, Rodilla V, Concheiro A, López-Castellano A, Alvarez-Lorenzo C. Micelles of Progesterone for Topical Eye Administration: Interspecies and Intertissues Differences in Ex Vivo Ocular Permeability. Pharmaceutics 2020; 12:pharmaceutics12080702. [PMID: 32722548 PMCID: PMC7464168 DOI: 10.3390/pharmaceutics12080702] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Revised: 07/14/2020] [Accepted: 07/23/2020] [Indexed: 01/04/2023] Open
Abstract
Progesterone (PG) may provide protection to the retina during retinitis pigmentosa, but its topical ocular supply is hampered by PG poor aqueous solubility and low ocular bioavailability. The development of efficient topical ocular forms must face up to two relevant challenges: Protective barriers of the eyes and lack of validated ex vivo tests to predict drug permeability. The aims of this study were: (i) To design micelles using Pluronic F68 and Soluplus copolymers to overcome PG solubility and permeability; and (ii) to compare drug diffusion through the cornea and sclera of three animal species (rabbit, porcine, and bovine) to investigate interspecies differences. Micelles of Pluronic F68 (3–4 nm) and Soluplus (52–59 nm) increased PG solubility by one and two orders of magnitude, respectively and exhibited nearly a 100% encapsulation efficiency. Soluplus systems showed in situ gelling capability in contrast to the low viscosity Pluronic F68 micelles. The formulations successfully passed the hen’s egg-chorioallantoic membrane test (HET-CAM) test. PG penetration through rabbit cornea and sclera was faster than through porcine or bovine cornea, although the differences were also formulation-dependent. Porcine tissues showed intermediate permeability between rabbit and bovine. Soluplus micelles allowed greater PG accumulation in cornea and sclera whereas Pluronic F68 promoted a faster penetration of lower PG doses.
Collapse
Affiliation(s)
- Adrián M. Alambiaga-Caravaca
- Departamento de Farmacia, Facultad de Ciencias de la Salud, Instituto de Ciencias Biomédicas, Universidad Cardenal Herrera-CEU, CEU Universities, C/Santiago Ramón y Cajal, s/n., Alfara del Patriarca, 46115 Valencia, Spain; (A.M.A.-C.); (M.A.C.-P.); (V.R.)
| | - María Aracely Calatayud-Pascual
- Departamento de Farmacia, Facultad de Ciencias de la Salud, Instituto de Ciencias Biomédicas, Universidad Cardenal Herrera-CEU, CEU Universities, C/Santiago Ramón y Cajal, s/n., Alfara del Patriarca, 46115 Valencia, Spain; (A.M.A.-C.); (M.A.C.-P.); (V.R.)
| | - Vicent Rodilla
- Departamento de Farmacia, Facultad de Ciencias de la Salud, Instituto de Ciencias Biomédicas, Universidad Cardenal Herrera-CEU, CEU Universities, C/Santiago Ramón y Cajal, s/n., Alfara del Patriarca, 46115 Valencia, Spain; (A.M.A.-C.); (M.A.C.-P.); (V.R.)
| | - Angel Concheiro
- Departamento de Farmacología, Farmacia y Tecnología Farmacéutica, I+D Farma Group, Facultad de Farmacia, and Health Research Institute of Santiago de Compostela (IDIS), Universidade de Santiago de Compostela, 15782 Santiago de Compostela, Spain;
| | - Alicia López-Castellano
- Departamento de Farmacia, Facultad de Ciencias de la Salud, Instituto de Ciencias Biomédicas, Universidad Cardenal Herrera-CEU, CEU Universities, C/Santiago Ramón y Cajal, s/n., Alfara del Patriarca, 46115 Valencia, Spain; (A.M.A.-C.); (M.A.C.-P.); (V.R.)
- Correspondence: (A.L.-C.); (C.A.-L.); Tel.: +34-961-369-000 (ext. 64906) (A.L.-C.); +34-881-815-239 (C.A.-L.)
| | - Carmen Alvarez-Lorenzo
- Departamento de Farmacología, Farmacia y Tecnología Farmacéutica, I+D Farma Group, Facultad de Farmacia, and Health Research Institute of Santiago de Compostela (IDIS), Universidade de Santiago de Compostela, 15782 Santiago de Compostela, Spain;
- Correspondence: (A.L.-C.); (C.A.-L.); Tel.: +34-961-369-000 (ext. 64906) (A.L.-C.); +34-881-815-239 (C.A.-L.)
| |
Collapse
|
14
|
Blair NP, Leahy S, Nathanael Matei, Shahidi M. Control of retinal blood flow levels by selected combinations of cervical arterial ligations in rat. Exp Eye Res 2020; 197:108088. [PMID: 32502531 DOI: 10.1016/j.exer.2020.108088] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Revised: 05/26/2020] [Accepted: 05/27/2020] [Indexed: 10/24/2022]
Abstract
The effect of various combinations of cervical arterial ligations (Combinations) on retinal blood flow (RBF) levels is not known in rats. We hypothesized: 1) No artery exists between the Circle of Willis and the eye, 2) Selective Combinations enable varying RBF levels between normal and zero, 3) In certain Combinations, the capillary bed of the head participates in supplying the eye. Twenty-six Combinations were studied in one eye of 20 Long-Evans rats under general anesthesia. RBF was quantitatively evaluated with our published imaging methods based on direct measurements of venous diameter and blood velocity from the displacement of fluorescent microspheres over time. For each Combination, one or more RBF values (runs) were measured. Data were obtained from 59 runs (2.9 ± 2.7 runs/rat). Levels of RBF ranged from normal to zero. An artery between the Circle of Willis and the eye was excluded. With some Combinations, flow traversed the capillary bed. Combinations were consolidated into five Groups based on the blood flow paths remaining after the ligations. A mixed linear model accounting for multiple measurements in the same eye demonstrated an effect of Group on RBF (P < 0.0005). By major source of ocular blood supply, the trend of RBF levels was: ipsilateral carotid artery > contralateral carotid artery > ipsilateral distal internal carotid artery retrograde from Circle of Willis. The findings advanced knowledge of the sources of blood supply to the rat eye and demonstrated a method of selective cervical arterial ligations for varying RBF levels with potential to impact future retinal ischemia research.
Collapse
Affiliation(s)
- Norman P Blair
- Department of Ophthalmology and Visual Sciences, University of Illinois at Chicago, United States.
| | - Sophie Leahy
- Department of Ophthalmology, University of Southern California, United States.
| | - Nathanael Matei
- Department of Ophthalmology, University of Southern California, United States.
| | - Mahnaz Shahidi
- Department of Ophthalmology, University of Southern California, United States.
| |
Collapse
|
15
|
Allen RS, Feola A, Motz CT, Ottensmeyer AL, Chesler KC, Dunn R, Thulé PM, Pardue MT. Retinal Deficits Precede Cognitive and Motor Deficits in a Rat Model of Type II Diabetes. Invest Ophthalmol Vis Sci 2019; 60:123-133. [PMID: 30640976 DOI: 10.1167/iovs.18-25110] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Purpose To investigate the temporal appearance of retinal, cognitive, and motor deficits in Goto-Kakizaki (GK) rats, a spontaneously occurring, polygenic model of type II diabetes. GK rats develop impaired insulin secretion at 2 weeks and fasting hyperglycemia at 4 weeks. Methods In male and female GK rats and Wistar controls, glucose tolerance test (hyperglycemia) and electroretinogram (ERG, retinal function) were performed at 4 and 8 weeks of age. Spectral domain-optical coherence tomography (retinal structure) was assessed at 6 weeks. Spatial alternation (cognitive function) and number of entries (exploratory behavior) were assessed via Y-maze at 4, 5, 6, 7, and 8 weeks. Rotarod (motor function) was performed at 4, 6, and 8 weeks. Results By 4 weeks, the GK rats exhibited significant glucose intolerance (P < 0.001) and retinal deficits, including delays in ERG implicit times (flicker, P < 0.01; oscillatory potentials, P < 0.001). In addition, the GK rats showed greater ERG amplitudes (P < 0.001) and thinner retinas (P < 0.001). At 7 weeks, the GK rats showed deficits in cognitive function (P < 0.001) and exploratory behavior (P < 0.01). However, no motor function deficits were observed by 8 weeks. Interestingly, the male GK rats showed greater hyperglycemia (P < 0.05), but the female rats showed greater ERG delays (P < 0.001). Conclusions In GK rats, retinal function deficits developed prior to cognitive or motor deficits. Future studies will investigate common mechanistic links, long-term functional and vascular changes, and whether early retinal deficits can predict cognitive dysfunction or late-stage retinal disease.
Collapse
Affiliation(s)
- Rachael S Allen
- Atlanta VA Center for Visual and Neurocognitive Rehabilitation, Atlanta VA Health Care System, Decatur, Georgia, United States.,Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, Georgia, United States
| | - Andrew Feola
- Atlanta VA Center for Visual and Neurocognitive Rehabilitation, Atlanta VA Health Care System, Decatur, Georgia, United States.,Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, Georgia, United States
| | - Cara T Motz
- Atlanta VA Center for Visual and Neurocognitive Rehabilitation, Atlanta VA Health Care System, Decatur, Georgia, United States
| | - Amy L Ottensmeyer
- Atlanta VA Center for Visual and Neurocognitive Rehabilitation, Atlanta VA Health Care System, Decatur, Georgia, United States.,Department of Ophthalmology, Emory University, Atlanta, Georgia, United States
| | - Kyle C Chesler
- Atlanta VA Center for Visual and Neurocognitive Rehabilitation, Atlanta VA Health Care System, Decatur, Georgia, United States.,Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, Georgia, United States
| | - Ryan Dunn
- Atlanta VA Center for Visual and Neurocognitive Rehabilitation, Atlanta VA Health Care System, Decatur, Georgia, United States.,Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, Georgia, United States
| | - Peter M Thulé
- Section Endocrinology & Metabolism, Atlanta VA Health Care System & Emory University School of Medicine, Decatur, Georgia, United States
| | - Machelle T Pardue
- Atlanta VA Center for Visual and Neurocognitive Rehabilitation, Atlanta VA Health Care System, Decatur, Georgia, United States.,Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, Georgia, United States
| |
Collapse
|
16
|
Wang YY, Niu RZ, Wang JD, Jin Y, Wang TH, Liu F. Establishment of brain ischemia model in tree shrew. Brain Res 2019; 1718:194-200. [PMID: 31077648 DOI: 10.1016/j.brainres.2019.05.011] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2019] [Revised: 05/06/2019] [Accepted: 05/07/2019] [Indexed: 02/05/2023]
Abstract
BACKGROUND Tree shrew, as a kind of small and inexpensive animal between insectivores and primates with the general anatomy being similar to human, could be considered as developed animal model for brain ischemia (BI) study. However, there is no neural behavior scores criterion from tree shrew with BI up to now. METHODS To produce BI model of tree shrew, a novel systematic neurobehavioral assessment scale, named as neural behavior scores (NBS) including aggressive behavior, seeking behavior, gait, startle reflex, high jump and warped-tail phenomenon was firstly established and used in this study. Moreover, magnetic resonance imaging (MRI) was performed on the first day after the operation to detect the imaging changes caused by ischemia. Then TTC, HE staining and immunofluorescent staining for GFAP and NeuN, were performed 24 h after surgery respectively. RESULTS NBS in BI group were significantly higher than that of sham operation group at 1d, 3d, 5d and 7d after ischemia. Meanwhile, compared with the sham operation group, the T2 images demonstrated significant higher signal and local brain swelling after cerebral ischemia in tree shrews. The staining of TTC and HE showed apparent infarction and necrosis of the cerebral region, and most of neurons exhibited a shrink. CONCLUSION We have successfully established the BI model of tree shrew, confirmed by NBS (a new developed method), MRI, HE staining, TTC staining and immunofluorescence staining. It is the first time to report a novel neurobehavioral assessment scale for BI in tree shrew.
Collapse
Affiliation(s)
- Yang-Yang Wang
- Institute of Neurological Disease, Department of Anesthesiology, Translational Neuroscience Center, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Rui-Ze Niu
- Laboratory Zoology Department, Institute of Neuroscience, Kunming Medical University, Kunming 650000, China
| | - Jie-Dong Wang
- Laboratory Zoology Department, Institute of Neuroscience, Kunming Medical University, Kunming 650000, China
| | - Yuan Jin
- Laboratory Zoology Department, Institute of Neuroscience, Kunming Medical University, Kunming 650000, China
| | - Ting-Hua Wang
- Institute of Neurological Disease, Department of Anesthesiology, Translational Neuroscience Center, West China Hospital, Sichuan University, Chengdu 610041, China; Laboratory Zoology Department, Institute of Neuroscience, Kunming Medical University, Kunming 650000, China.
| | - Fei Liu
- Institute of Neurological Disease, Department of Anesthesiology, Translational Neuroscience Center, West China Hospital, Sichuan University, Chengdu 610041, China.
| |
Collapse
|
17
|
Progesterone, Lipoic Acid, and Sulforaphane as Promising Antioxidants for Retinal Diseases: A Review. Antioxidants (Basel) 2019; 8:antiox8030053. [PMID: 30832304 PMCID: PMC6466531 DOI: 10.3390/antiox8030053] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2019] [Revised: 02/21/2019] [Accepted: 02/28/2019] [Indexed: 12/19/2022] Open
Abstract
Oxidative stress has been documented to be a key factor in the cause and progression of different retinal diseases. Oxidative cellular unbalance triggers a sequence of reactions which prompt cell degeneration and retinal dysfunction, both hallmarks of several retinal pathologies. There is no effective treatment, yet, for many retinal diseases. Antioxidant treatment have been pointed out to be an encouraging palliative treatment; the beneficial effects documented involve slowing the progression of the disease, a reduction of cell degeneration, and improvement of retinal functions. There is a vast information corpus on antioxidant candidates. In this review, we expose three of the main antioxidant treatments, selected for their promising results that has been reported to date. Recently, the sulforaphane, an isothiocyanate molecule, has been unveiled as a neuroprotective candidate, by its antioxidant properties. Progesterone, a neurosteroid has been proposed to be a solid and effective neuroprotective agent. Finally, the lipoic acid, an organosulfur compound, is a well-recognized antioxidant. All of them, have been tested and studied on different retinal disease models. In this review, we summarized the published results of these works, to offer a general view of the current antioxidant treatment advances, including the main effects and mechanisms described.
Collapse
|
18
|
Allen RS, Motz CT, Feola A, Chesler KC, Haider R, Ramachandra Rao S, Skelton LA, Fliesler SJ, Pardue MT. Long-Term Functional and Structural Consequences of Primary Blast Overpressure to the Eye. J Neurotrauma 2018; 35:2104-2116. [PMID: 29648979 DOI: 10.1089/neu.2017.5394] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Acoustic blast overpressure (ABO) injury in military personnel and civilians is often accompanied by delayed visual deficits. However, most animal model studies dealing with blast-induced visual defects have focused on short-term (≤1 month) changes. Here, we evaluated long-term (≤8 months) retinal structure and function deficits in rats with ABO injury. Adult male Long-Evans rats were subjected to ABO from a single blast (approximately 190 dB SPL, ∼63 kPa, @80 psi), generated by a shock tube device. Retinal function (electroretinography; ERG), visual function (optomotor response), retinal thickness (spectral domain-optical coherence tomography; SD-OCT), and spatial cognition/exploratory motor behavior (Y-maze) were measured at 2, 4, 6, and 8 months post-blast. Immunohistochemical analysis of glial fibrillary acidic protein (GFAP) in retinal sections was performed at 8 months post-blast. Electroretinogram a- and b-waves, oscillatory potentials, and flicker responses showed greater amplitudes with delayed implicit times in both eyes of blast-exposed animals, relative to controls. Contrast sensitivity (CS) was reduced in both eyes of blast-exposed animals, whereas spatial frequency (SF) was decreased only in ipsilateral eyes, relative to controls. Total retinal thickness was greater in both eyes of blast-exposed animals, relative to controls, due to increased thickness of several retinal layers. Age, but not blast exposure, altered Y-maze outcomes. GFAP was greatly increased in blast-exposed retinas. ABO exposure resulted in visual and retinal changes that persisted up to 8 months post-blast, mimicking some of the visual deficits observed in human blast-exposed patients, thereby making this a useful model to study mechanisms of injury and potential treatments.
Collapse
Affiliation(s)
- Rachael S Allen
- 1 Center for Visual and Neurocognitive Rehabilitation, Atlanta VA Medical Center , Atlanta, Georgia .,2 Biomedical Engineering, Georgia Institute of Technology , Atlanta, Georgia
| | - Cara T Motz
- 1 Center for Visual and Neurocognitive Rehabilitation, Atlanta VA Medical Center , Atlanta, Georgia
| | - Andrew Feola
- 1 Center for Visual and Neurocognitive Rehabilitation, Atlanta VA Medical Center , Atlanta, Georgia .,2 Biomedical Engineering, Georgia Institute of Technology , Atlanta, Georgia
| | - Kyle C Chesler
- 2 Biomedical Engineering, Georgia Institute of Technology , Atlanta, Georgia
| | - Raza Haider
- 1 Center for Visual and Neurocognitive Rehabilitation, Atlanta VA Medical Center , Atlanta, Georgia
| | - Sriganesh Ramachandra Rao
- 3 Ophthalmology, Biochemistry, and Neuroscience Program, SUNY-University at Buffalo , Buffalo, New York
| | - Lara A Skelton
- 4 Research Service, VA Western NY Healthcare System , Buffalo, New York
| | - Steven J Fliesler
- 3 Ophthalmology, Biochemistry, and Neuroscience Program, SUNY-University at Buffalo , Buffalo, New York.,4 Research Service, VA Western NY Healthcare System , Buffalo, New York
| | - Machelle T Pardue
- 1 Center for Visual and Neurocognitive Rehabilitation, Atlanta VA Medical Center , Atlanta, Georgia .,2 Biomedical Engineering, Georgia Institute of Technology , Atlanta, Georgia
| |
Collapse
|
19
|
Pardue MT, Allen RS. Neuroprotective strategies for retinal disease. Prog Retin Eye Res 2018; 65:50-76. [PMID: 29481975 PMCID: PMC6081194 DOI: 10.1016/j.preteyeres.2018.02.002] [Citation(s) in RCA: 158] [Impact Index Per Article: 26.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2017] [Revised: 02/14/2018] [Accepted: 02/20/2018] [Indexed: 12/20/2022]
Abstract
Diseases that affect the eye, including photoreceptor degeneration, diabetic retinopathy, and glaucoma, affect 11.8 million people in the US, resulting in vision loss and blindness. Loss of sight affects patient quality of life and puts an economic burden both on individuals and the greater healthcare system. Despite the urgent need for treatments, few effective options currently exist in the clinic. Here, we review research on promising neuroprotective strategies that promote neuronal survival with the potential to protect against vision loss and retinal cell death. Due to the large number of neuroprotective strategies, we restricted our review to approaches that we had direct experience with in the laboratory. We focus on drugs that target survival pathways, including bile acids like UDCA and TUDCA, steroid hormones like progesterone, therapies that target retinal dopamine, and neurotrophic factors. In addition, we review rehabilitative methods that increase endogenous repair mechanisms, including exercise and electrical stimulation therapies. For each approach, we provide background on the neuroprotective strategy, including history of use in other diseases; describe potential mechanisms of action; review the body of research performed in the retina thus far, both in animals and in humans; and discuss considerations when translating each treatment to the clinic and to the retina, including which therapies show the most promise for each retinal disease. Despite the high incidence of retinal diseases and the complexity of mechanisms involved, several promising neuroprotective treatments provide hope to prevent blindness. We discuss attractive candidates here with the goal of furthering retinal research in critical areas to rapidly translate neuroprotective strategies into the clinic.
Collapse
Affiliation(s)
- Machelle T Pardue
- Center for Visual and Neurocognitive Rehabilitation, Atlanta VA Medical Center, 1670 Clairmont Road, Decatur, GA, 30033, USA; Department of Biomedical Engineering, Georgia Institute of Technology, 313 Ferst Drive, Atlanta, GA, 30332, USA.
| | - Rachael S Allen
- Center for Visual and Neurocognitive Rehabilitation, Atlanta VA Medical Center, 1670 Clairmont Road, Decatur, GA, 30033, USA
| |
Collapse
|
20
|
Güleç Başer B, İslimye Taşkın M, Adalı E, Öztürk E, Hısmıoğulları AA, Yay A. Does progesterone have protective effects on ovarian ischemia-reperfusion injury? J Turk Ger Gynecol Assoc 2018; 19:87-93. [PMID: 29545230 PMCID: PMC5994815 DOI: 10.4274/jtgga.2017.0047] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
Objective: The aim of the present study was to evaluate the effects of progesterone (PG) against ovarian ischemia-reperfusion (I/R) injury through the evaluation of biochemical and histopathologic parameters. Material and Methods: Twenty-one female Wistar albino rats were divided into three groups. Group 1: Sham; group 2: I/R; group 3: I/R+PG (8 mg/kg). PG was administered intraperitoneally to the rats in group 3, 30 minutes before a detorsion operation. Ovarian I/R injury was evaluated in serum and tissue by using biochemical parameters including malondialdehyde (MDA), total antioxidant status (TAS), total oxidant status (TOS), oxidative stress index, neutrophil gelatinase-associated lipocalin (NGAL) and immunofluorescence staining by using a terminal deoxynucleotidyl transferase-mediated dUTP nick end labeling (TUNEL) assay. Results: Serum and tissue TOS levels were significantly lower in group 3 than in group 2. Tissue TAS levels were higher in group 3 than in group 2 (p<0.001). NGAL and MDA levels were similar between the groups. Histologic score, including vascular congestion, hemorrhage, polymorphonuclear neutrophils, and interstitial edema, was higher in group 2. Pre-treatment with PG decreased the score, but this difference was not statistically significant. The number of apoptotic cells was higher in group 2 than in groups 1 and 3. The TUNEL-positive cell number decreased with PG in group 3. Conclusion: Preoperative PG treatment might exert protective effects on ovarian I/R injury through its anti-apoptotic and antioxidative properties.
Collapse
Affiliation(s)
- Banu Güleç Başer
- Department of Obstetrics and Gynecology, Balıkesir University School of Medicine, Balıkesir, Turkey
| | - Mine İslimye Taşkın
- Department of Obstetrics and Gynecology, Balıkesir University School of Medicine, Balıkesir, Turkey
| | - Ertan Adalı
- Department of Obstetrics and Gynecology, Balıkesir University School of Medicine, Balıkesir, Turkey
| | - Emine Öztürk
- Department of Obstetrics and Gynecology, Balıkesir University School of Medicine, Balıkesir, Turkey
| | | | - Arzu Yay
- Department of Histology and Embryology, Erciyes University School of Medicine, Kayseri, Turkey
| |
Collapse
|
21
|
Ramírez-Lamelas DT, Benlloch-Navarro S, López-Pedrajas R, Gimeno-Hernández R, Olivar T, Silvestre D, Miranda M. Lipoic Acid and Progesterone Alone or in Combination Ameliorate Retinal Degeneration in an Experimental Model of Hereditary Retinal Degeneration. Front Pharmacol 2018; 9:469. [PMID: 29867476 PMCID: PMC5954235 DOI: 10.3389/fphar.2018.00469] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2018] [Accepted: 04/23/2018] [Indexed: 01/27/2023] Open
Abstract
Retinitis pigmentosa (RP) is a group of inherited retinopathies characterized by photoreceptors death. Our group has shown the positive progesterone (P4) actions on cell death progression in an experimental model of RP. In an effort to enhance the beneficial effects of P4, the aim of this study was to combine P4 treatment with an antioxidant [lipoic acid (LA)] in the rd1 mice. rd1 and control mice were treated with 100 mg/kg body weight of P4, LA, or a combination of both on postnatal day 7 (PN7), 9, and 11, and were sacrificed at PN11. The administration of LA and/or P4 diminishes cell death in rd1 retinas. The effect obtained after the combined administration of LA and P4 is higher than the one obtained with LA or P4 alone. The three treatments decreased GFAP staining, however, in the far peripheral retina, and the two treatments that offered better results were LA and LA plus P4. LA or LA plus P4 increased retinal glutathione (GSH) concentration in the rd1 mice. Although LA and P4 are able to protect photoreceptors from death in rd1 mice retinas, a better effectiveness is achieved when administering LA and P4 at the same time.
Collapse
Affiliation(s)
- Dolores T Ramírez-Lamelas
- Departamento Ciencias Biomédicas, Universidad CEU Cardenal Herrera, CEU Universities, Valencia, Spain
| | - Soledad Benlloch-Navarro
- Departamento Ciencias Biomédicas, Universidad CEU Cardenal Herrera, CEU Universities, Valencia, Spain
| | - Rosa López-Pedrajas
- Departamento Ciencias Biomédicas, Universidad CEU Cardenal Herrera, CEU Universities, Valencia, Spain.,Instituto de Ciencias Biomédicas, Universidad CEU Cardenal Herrera, CEU Universities, Valencia, Spain
| | - Roberto Gimeno-Hernández
- Departamento Ciencias Biomédicas, Universidad CEU Cardenal Herrera, CEU Universities, Valencia, Spain
| | - Teresa Olivar
- Departamento Ciencias Biomédicas, Universidad CEU Cardenal Herrera, CEU Universities, Valencia, Spain
| | - Dolores Silvestre
- Departamento Farmacia, Universidad CEU Cardenal Herrera, CEU Universities, Valencia, Spain
| | - María Miranda
- Departamento Ciencias Biomédicas, Universidad CEU Cardenal Herrera, CEU Universities, Valencia, Spain.,Instituto de Ciencias Biomédicas, Universidad CEU Cardenal Herrera, CEU Universities, Valencia, Spain
| |
Collapse
|
22
|
Sergeeva EG, Espinosa-Garcia C, Atif F, Pardue MT, Stein DG. Neurosteroid allopregnanolone reduces ipsilateral visual cortex potentiation following unilateral optic nerve injury. Exp Neurol 2018; 306:138-148. [PMID: 29729249 DOI: 10.1016/j.expneurol.2018.05.005] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2018] [Revised: 04/06/2018] [Accepted: 05/01/2018] [Indexed: 10/17/2022]
Abstract
In adult mice with unilateral optic nerve crush injury (ONC), we studied visual response plasticity in the visual cortex following stimulation with sinusoidal grating. We examined visually evoked potentials (VEP) in the primary visual cortex ipsilateral and contralateral to the crushed nerve. We found that unilateral ONC induces enhancement of visual response on the side ipsilateral to the injury that is evoked by visual stimulation to the intact eye. This enhancement was associated with supranormal spatial frequency thresholds in the intact eye when tested using optomotor response. To probe whether injury-induced disinhibition caused the potentiation, we treated animals with the neurosteroid allopregnanolone, a potent agonist of the GABAA receptor, one hour after crush and on post-injury days 3, 8, 13, and 18. Allopregnanolone diminished enhancement of the VEP and this effect was associated with the upregulated synthesis of the δ-subunit of the GABAA receptor. Our study shows a new aspect of experience-dependent plasticity following unilateral ONC. This hyper-activity in the ipsilateral visual cortex is prevented by upregulation of GABA inhibition with allopregnanolone. Our findings suggest the therapeutic potential of allopregnanolone for modulation of plasticity in certain eye and brain disorders and a possible role for disinhibition in ipsilateral hyper-activity following unilateral ONC.
Collapse
Affiliation(s)
- Elena G Sergeeva
- Department of Emergency Medicine, Emory University, 1365B Clifton Road NE, Suite 5100, Atlanta, GA 30322, USA.
| | - Claudia Espinosa-Garcia
- Department of Emergency Medicine, Emory University, 1365B Clifton Road NE, Suite 5100, Atlanta, GA 30322, USA
| | - Fahim Atif
- Department of Emergency Medicine, Emory University, 1365B Clifton Road NE, Suite 5100, Atlanta, GA 30322, USA
| | - Machelle T Pardue
- Center for Visual and Neurocognitive Rehabilitation, Atlanta VA Medical Center, 1670 Clairmont Road, Decatur, GA 30033, USA; Department of Biomedical Engineering, Georgia Institute of Technology, 313 Ferst Drive, Atlanta, GA 30332, USA
| | - Donald G Stein
- Department of Emergency Medicine, Emory University, 1365B Clifton Road NE, Suite 5100, Atlanta, GA 30322, USA.
| |
Collapse
|
23
|
Allen RS, Sayeed I, Oumarbaeva Y, Morrison KC, Choi PH, Pardue MT, Stein DG. Progesterone treatment shows greater protection in brain vs. retina in a rat model of middle cerebral artery occlusion: Progesterone receptor levels may play an important role. Restor Neurol Neurosci 2018; 34:947-963. [PMID: 27802245 DOI: 10.3233/rnn-160672] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
BACKGROUND/OBJECTIVE To determine whether inflammation increases in retina as it does in brain following middle cerebral artery occlusion (MCAO), and whether the neurosteroid progesterone, shown to have protective effects in both retina and brain after MCAO, reduces inflammation in retina as well as brain. METHODS MCAO rats treated systemically with progesterone or vehicle were compared with shams. Protein levels of cytosolic NF-κB, nuclear NF-κB, phosphorylated NF-κB, IL-6, TNF-α, CD11b, progesterone receptor A and B, and pregnane X receptor were assessed in retinas and brains at 24 and 48 h using western blots. RESULTS Following MCAO, significant increases were observed in the following inflammatory markers: pNF-κB and CD11b at 24 h in both brain and retina, nuclear NF-κB at 24 h in brain and 48 h in retina, and TNF-α at 24 h in brain.Progesterone treatment in MCAO animals significantly attenuated levels of the following markers in brain: pNF-κB, nuclear NF-κB, IL-6, TNF-α, and CD11b, with significantly increased levels of cytosolic NF-κB. Retinas from progesterone-treated animals showed significantly reduced levels of nuclear NF-κB and IL-6 and increased levels of cytosolic NF-κB, with a trend for reduction in other markers. Post-MCAO, progesterone receptors A and B were upregulated in brain and downregulated in retina. CONCLUSION Inflammatory markers increased in both brain and retina after MCAO, with greater increases observed in brain. Progesterone treatment reduced inflammation, with more dramatic reductions observed in brain than retina. This differential effect may be due to differences in the response of progesterone receptors in brain and retina after injury.
Collapse
Affiliation(s)
- Rachael S Allen
- Department of Emergency Medicine, Emory University, Atlanta, GA, USA.,Department of Ophthalmology, Emory University, Atlanta, GA, USA.,Center for Visual and Neurocognitive Rehabilitation, Atlanta VA Medical Center, Decatur, GA, USA
| | - Iqbal Sayeed
- Department of Emergency Medicine, Emory University, Atlanta, GA, USA
| | - Yuliya Oumarbaeva
- Department of Emergency Medicine, Emory University, Atlanta, GA, USA
| | | | - Paul H Choi
- Department of Emergency Medicine, Emory University, Atlanta, GA, USA
| | - Machelle T Pardue
- Department of Ophthalmology, Emory University, Atlanta, GA, USA.,Center for Visual and Neurocognitive Rehabilitation, Atlanta VA Medical Center, Decatur, GA, USA
| | - Donald G Stein
- Department of Emergency Medicine, Emory University, Atlanta, GA, USA
| |
Collapse
|
24
|
Chen X, Chen C, Hao J, Zhang J, Zhang F. Effect of CLIP3 Upregulation on Astrocyte Proliferation and Subsequent Glial Scar Formation in the Rat Spinal Cord via STAT3 Pathway After Injury. J Mol Neurosci 2017; 64:117-128. [PMID: 29218499 DOI: 10.1007/s12031-017-0998-6] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2017] [Accepted: 11/06/2017] [Indexed: 12/16/2022]
Abstract
Spinal cord injury (SCI) is a devastating event resulting in neuron degeneration and permanent paralysis through inflammatory cytokine overproduction and glial scar formation. Presently, the endogenous molecular mechanisms coordinating glial scar formation in the injured spinal cord remain elusive. Signal transducer and activator of transcription 3 (STAT3) is a well-known transcription factor particularly involving in cell proliferation and inflammation in the lesion site following SCI. Meanwhile, CAP-Gly domain containing linker protein 3(CLIP3), a vital cytoplasmic protein, has been confirmed to providing an optimal conduit for intracellular signal transduction and interacting with STAT3 with mass spectrometry analysis. In this study, we aimed to identify the expression of CLIP3 in the spinal cord as well as its role in mediating astrocyte activation and glial scar formation after SCI by establishing an acute traumatic SCI model in male adult rats. Western blot analysis revealed that CLIP3 increased gradually after injury, reached a peak at day 3. The immunohistochemistry staining showed the same result in white matter. With double immunofluorescence staining, we found that CLIP3 was expressed in glial cells and significant changes of CLIP3 expression occurred in astrocytes during the pathological process. Statistical analysis demonstrated there was a correlation between the number of positive cells stained by CLIP3 and STAT3 in the spinal cord after SCI. Co-immunoprecipitation further indicated that CLIP3 interacted with STAT3 in the injured spinal cord. Taken together, our study clearly suggested that CLIP3 played an essential role in astrocyte activation, associating with the STAT3 pathway activation induced by SCI.
Collapse
Affiliation(s)
- Xiaoqing Chen
- Department of Orthopedics, Affiliated Hospital of Nantong University, Nantong, Jiangsu, 226001, China.,Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Nantong, Jiangsu, 226001, China
| | - Cheng Chen
- Department of Orthopedics, Affiliated Hospital of Nantong University, Nantong, Jiangsu, 226001, China.,Medical Colleges of Nantong University, Nantong, Jiangsu, 226001, China
| | - Jie Hao
- Department of Orthopedics, Affiliated Hospital of Nantong University, Nantong, Jiangsu, 226001, China.,Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Nantong, Jiangsu, 226001, China
| | - Jiyun Zhang
- Medical Colleges of Nantong University, Nantong, Jiangsu, 226001, China.,Department of Radiology, Third Municipal People's Hospital, Nantong, Jiangsu, 226001, China
| | - Feng Zhang
- Department of Orthopedics, Affiliated Hospital of Nantong University, Nantong, Jiangsu, 226001, China. .,Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Nantong, Jiangsu, 226001, China.
| |
Collapse
|
25
|
Effect of Progesterone on Cerebral Vasospasm and Neurobehavioral Outcomes in a Rodent Model of Subarachnoid Hemorrhage. World Neurosurg 2017; 110:e150-e159. [PMID: 29097330 DOI: 10.1016/j.wneu.2017.10.118] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2017] [Revised: 10/19/2017] [Accepted: 10/21/2017] [Indexed: 11/20/2022]
Abstract
BACKGROUND Subarachnoid hemorrhage (SAH) induces widespread inflammation leading to cellular injury, vasospasm, and ischemia. Evidence suggests that progesterone (PROG) can improve functional recovery in acute brain injury owing to its anti-inflammatory and neuroprotective properties, which could also be beneficial in SAH. We hypothesized that PROG treatment attenuates inflammation-mediated cerebral vasospasm and microglial activation, improves synaptic connectivity, and ameliorates functional recovery after SAH. METHODS We investigated the effect of PROG in a cisternal SAH model in adult male C57BL/6 mice. Neurobehavioral outcomes were evaluated using rotarod latency and grip strength tests. Basilar artery perimeter, α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid glutamate receptor 1 (GluR1)/synaptophysin colocalization, and Iba-1 immunoreactivity were quantified histologically. RESULTS PROG (8 mg/kg) significantly improved rotarod latency at day 6 and grip strength at day 9. PROG-treated mice had significantly reduced basilar artery vasospasm at 24 hours. GluR1/synaptophysin colocalization, indicative of synaptic GluR1, was significantly reduced in the SAH+Vehicle group at 24 hours, and PROG treatment significantly attenuated this reduction. PROG treatment significantly reduced microglial cell activation and proliferation in cerebellum and cortex but not in the brainstem at 10 days. CONCLUSIONS PROG treatment ameliorated cerebral vasospasm, reduced microglial activation, restored synaptic GluR1 localization, and improved neurobehavioral performance in a murine model of SAH. These results provide a rationale for further translational testing of PROG therapy in SAH.
Collapse
|
26
|
Dai J, He J, Wang G, Wang M, Li S, Yin ZQ. Contribution of GABAa, GABAc and glycine receptors to rat dark-adapted oscillatory potentials in the time and frequency domain. Oncotarget 2017; 8:77696-77709. [PMID: 29100418 PMCID: PMC5652335 DOI: 10.18632/oncotarget.20770] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2017] [Accepted: 07/29/2017] [Indexed: 02/02/2023] Open
Abstract
Retinal oscillatory potentials (OPs) consist of a series of relatively high-frequency rhythmic wavelets, superimposed onto the ascending phase of the b-wave of the electroretinogram (ERG). However, the origin of OPs is uncertain and methods of measurement of OPs are diverse. In this study, we first isolated OPs from the rat ERG and fitted them with Gabor functions and found that the envelope of the OP contained information about maximum amplitude and time-to-peak to enable satisfactory quantification of the later OPs. And the OP/b-wave ratio should be evaluated to exclude an effect of the b-wave on the OPs. Next, we recorded OPs after intravitreal injection of 2-amino-4-phosphonobutyric acid (APB), tetrodotoxin (TTX), γ-aminobutyric acid (GABA), strychnine (STR), SR95531 (SR), isoguvacine (ISO), (1,2,5,6-tetrahydropyridin-4-yl) methylphosphinic acid (TPMPA) and GABA+TPMPA. We showed that GABA and APB only removed the later OPs, when compared to control eyes. TTX delayed the peak time, and STR, SR and ISO reduced the amplitude of OPs. TPMPA delayed the peak time but increased the ratio of OPs to b-wave. Furthermore, administration of combined GABA and TPMPA caused the later OPs to increase in amplitude with time, compared with those after delivery of GABA alone. Finally, we observed that GABAc and glycine receptors contributed to a low-frequency component of the OPs, while GABAa contributed to both components. These results suggest that the early components of the OPs are mainly generated by the photoreceptors, whilst the later components are mainly regulated by GABAa, GABAc and glycine receptors.
Collapse
Affiliation(s)
- Jiaman Dai
- College of Bioengineering, Chongqing University, Chongqing 400030, China.,Key Lab of Visual Damage and Regeneration & Restoration of Chongqing, Chongqing 400038, China
| | - Juncai He
- Key Lab of Visual Damage and Regeneration & Restoration of Chongqing, Chongqing 400038, China.,Southwest Hospital/Southwest Eye Hospital, Third Military Medical University, Chongqing 400038, China
| | - Gang Wang
- Key Lab of Visual Damage and Regeneration & Restoration of Chongqing, Chongqing 400038, China.,Southwest Hospital/Southwest Eye Hospital, Third Military Medical University, Chongqing 400038, China
| | - Min Wang
- Key Lab of Visual Damage and Regeneration & Restoration of Chongqing, Chongqing 400038, China.,Southwest Hospital/Southwest Eye Hospital, Third Military Medical University, Chongqing 400038, China
| | - Shiying Li
- Key Lab of Visual Damage and Regeneration & Restoration of Chongqing, Chongqing 400038, China.,Southwest Hospital/Southwest Eye Hospital, Third Military Medical University, Chongqing 400038, China
| | - Zheng Qin Yin
- College of Bioengineering, Chongqing University, Chongqing 400030, China.,Key Lab of Visual Damage and Regeneration & Restoration of Chongqing, Chongqing 400038, China.,Southwest Hospital/Southwest Eye Hospital, Third Military Medical University, Chongqing 400038, China
| |
Collapse
|
27
|
Wyse-Jackson AC, Roche SL, Ruiz-Lopez AM, Moloney JN, Byrne AM, Cotter TG. Progesterone analogue protects stressed photoreceptors via bFGF-mediated calcium influx. Eur J Neurosci 2016; 44:3067-3079. [PMID: 27763693 DOI: 10.1111/ejn.13445] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2016] [Revised: 10/14/2016] [Accepted: 10/17/2016] [Indexed: 01/16/2023]
Abstract
Retinitis pigmentosa (RP) is a degenerative retinal disease leading to photoreceptor cell loss. In 2011, our group identified the synthetic progesterone 'Norgestrel' as a potential treatment for RP. Subsequent research showed Norgestrel to work through progesterone receptor membrane component 1 (PGRMC1) activation and upregulation of neuroprotective basic fibroblast growth factor (bFGF). Using trophic factor deprivation of 661W photoreceptor-like cells, we aimed to further elucidate the mechanism leading to Norgestrel-induced neuroprotection. In the present manuscript, we show by flow cytometry and live-cell immunofluorescence that Norgestrel induces an increase in cytosolic calcium in both healthy and stressed 661Ws over 24 h. Specific PGRMC1 inhibition by AG205 (1 μm) showed this rise to be PGRMC1-dependent, primarily utilizing calcium from extracellular sources, for blockade of L-type calcium channels by verapamil (50 μm) prevented a Norgestrel-induced calcium influx in stressed cells. Calcium influx was also shown to be bFGF-dependent, for siRNA knock down of bFGF prevented Norgestrel-PGRMC1 induced changes in cytosolic calcium. Notably, we demonstrate PGRMC1-activation is necessary for Norgestrel-induced bFGF upregulation. We propose that Norgestrel protects through the following pathway: binding to and activating PGRMC1 expressed on the surface of photoreceptor cells, PGRMC1 activation drives bFGF upregulation and subsequent calcium influx. Importantly, raised intracellular calcium is critical to Norgestrel's protective efficacy, for extracellular calcium chelation by EGTA abrogates the protective effects of Norgestrel on stressed 661W cells in vitro.
Collapse
Affiliation(s)
- Alice C Wyse-Jackson
- Cell Development and Disease Laboratory, Biochemistry Department, Bioscience Research Institute, University College Cork, Western Road, Cork, Ireland
| | - Sarah L Roche
- Cell Development and Disease Laboratory, Biochemistry Department, Bioscience Research Institute, University College Cork, Western Road, Cork, Ireland
| | - Ana M Ruiz-Lopez
- Cell Development and Disease Laboratory, Biochemistry Department, Bioscience Research Institute, University College Cork, Western Road, Cork, Ireland
| | - Jennifer N Moloney
- Cell Development and Disease Laboratory, Biochemistry Department, Bioscience Research Institute, University College Cork, Western Road, Cork, Ireland
| | - Ashleigh M Byrne
- Cell Development and Disease Laboratory, Biochemistry Department, Bioscience Research Institute, University College Cork, Western Road, Cork, Ireland
| | - Thomas G Cotter
- Cell Development and Disease Laboratory, Biochemistry Department, Bioscience Research Institute, University College Cork, Western Road, Cork, Ireland
| |
Collapse
|