1
|
Moon TK, Kang IK, Lee KJ, Kim JH, Kim HJ, Han AR, Woo HN, Lee JY, Choi JS, Park K, Lee H. mTOR downregulation promotes anti-inflammatory responses via the CCL3-CCR5 axis in hypoxic retinopathy. Mol Ther Methods Clin Dev 2025; 33:101404. [PMID: 39897639 PMCID: PMC11787640 DOI: 10.1016/j.omtm.2024.101404] [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: 06/27/2024] [Accepted: 12/28/2024] [Indexed: 02/04/2025]
Abstract
Hypoxic retinopathies, including diabetic retinopathy, are major contributors to vision impairment, mainly due to accelerated angiogenesis and inflammation. Previously, we demonstrated that AAV2-shmTOR, effective across distinct species, holds therapeutic promise by modulating the activated mTOR pathway, yet its mechanisms for reducing inflammation remain largely unexplored. To investigate AAV2-shmTOR's impact on atypical inflammation in these conditions, we employed an in vivo model of oxygen-induced retinopathy and an in vitro model using rMC1 Müller cells. AAV2-shmTOR notably decreased mTOR expression in rMC1 cells under hypoxic conditions, as verified by co-staining for mTOR and glial fibrillary acidic protein (GFAP). It effectively interrupted the activation of mTOR signaling triggered by hypoxia. It diminished the secretion of CCL3 from rMC1 cells, consequently reducing microglial migration in response to conditioned media from AAV2-shmTOR-treated rMC1 cells. Notably, the virus lowered CCL3 expression in Müller cells and reduced the presence of CCR5-positive microglia in vivo, indicating its effectiveness in targeted inflammation management via the CCL3-CCR5 pathway. These findings thus highlight the potential of AAV2-shmTOR to exert anti-inflammatory effects by influencing the mTOR and subsequent CCL3-CCR5 pathways in hypoxic retinopathies, presenting a novel therapeutic approach for retinal diseases marked by hypoxia-driven inflammation.
Collapse
Affiliation(s)
- Tae Kwon Moon
- Asan Medical Institute of Convergence Science and Technology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
- Department of Microbiology, University of Ulsan, College of Medicine, Seoul, Republic of Korea
| | - Im Kyeung Kang
- Asan Medical Institute of Convergence Science and Technology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
- Department of Microbiology, University of Ulsan, College of Medicine, Seoul, Republic of Korea
| | - Kyoung Jin Lee
- Department of Microbiology, University of Ulsan, College of Medicine, Seoul, Republic of Korea
- Bio-Medical Institute of Technology, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Ji Hyun Kim
- Department of Microbiology, University of Ulsan, College of Medicine, Seoul, Republic of Korea
- Bio-Medical Institute of Technology, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Hee Jong Kim
- Institute of New Drug Development Research, CdmoGen Co., Ltd., Seoul 05854, Republic of Korea
| | - A. Reum Han
- Asan Medical Institute of Convergence Science and Technology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
- Department of Biochemistry & Molecular Biology, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Ha-Na Woo
- Bio-Medical Institute of Technology, University of Ulsan College of Medicine, Seoul, Republic of Korea
- Department of Biochemistry & Molecular Biology, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Joo Yong Lee
- Bio-Medical Institute of Technology, University of Ulsan College of Medicine, Seoul, Republic of Korea
- Department of Ophthalmology, Asan Medical Center, College of Medicine, University of Ulsan, Seoul, Republic of Korea
| | - Jun-Sub Choi
- Institute of New Drug Development Research, CdmoGen Co., Ltd., Seoul 05854, Republic of Korea
| | - Keerang Park
- Institute of New Drug Development Research, CdmoGen Co., Ltd., Seoul 05854, Republic of Korea
| | - Heuiran Lee
- Bio-Medical Institute of Technology, University of Ulsan College of Medicine, Seoul, Republic of Korea
- Department of Microbiology, Asan Medical Center, College of Medicine, University of Ulsan, Seoul, Republic of Korea
| |
Collapse
|
2
|
Hanna J, Touahri Y, Pak A, David LA, van Oosten E, Dixit R, Vecchio LM, Mehta DN, Minamisono R, Aubert I, Schuurmans C. Pten Loss Triggers Progressive Photoreceptor Degeneration in an mTORC1-Independent Manner. Invest Ophthalmol Vis Sci 2025; 66:45. [PMID: 40116678 PMCID: PMC11935561 DOI: 10.1167/iovs.66.3.45] [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] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Accepted: 02/19/2025] [Indexed: 03/23/2025] Open
Abstract
Purpose Silencing Phosphatase and tensin homolog (Pten) is a proposed therapeutic strategy for tissue regeneration to treat neurological disorders. However, Pten is pleiotropic, inhibiting several signaling and metabolic pathways, including mTORC1 and glycolysis, both pro-regenerative in certain contexts. This study aims to assess the long-term impact of inactivating Pten on photoreceptor survival in the retina and to identify downstream pathway(s). Methods We assessed retinal integrity in Pten conditional knock-outs (cKOs) that were retinal progenitor cell (RPC)-specific (Pten RPC-cKO), a congenital model, or rod-specific (Pten Rho-cKO). We examined early changes in photoreceptor gene expression and used immunostaining to assess photoreceptors, reactive astrocytes, microglia, angiogenesis, and subretinal deposit formation from postnatal day (P) 21 to 1 year of age. Pten RPC-cKO retinal explants were treated with rapamycin, an mTOR inhibitor, or 2-deoxy-D-glucose (2DG), a glycolysis inhibitor. Results In both Pten-cKO models, retinas display signs of early pathogenesis as photoreceptor-specific gene expression is downregulated at P0, before photoreceptor loss. Pten loss triggers progressive rod and cone degeneration beginning at P21 in Pten RPC-cKOs and at 6 months of age in Pten Rho-cKOs. Activated microglia and astrocytes, and increased angiogenesis, are observed in both Pten-cKO models, while subretinal amyloid-β deposits develop in Pten RPC-cKOs. Rapamycin accelerates photoreceptor degeneration in Pten RPC-cKOs, whereas 2DG has no effect. Conclusions Our findings suggest that Pten loss, either in RPCs as a congenital model, or solely in mature rod photoreceptors, leads to progressive retinal degeneration that is exacerbated by mTORC1 suppression, drawing into question the therapeutic value of Pten-mTORC1 manipulations.
Collapse
Affiliation(s)
- Joseph Hanna
- Biological Sciences Platform, Sunnybrook Research Institute, Toronto, Ontario, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
- Department of Ophthalmology and Vision Sciences, University of Toronto, Toronto, Ontario, Canada
| | - Yacine Touahri
- Biological Sciences Platform, Sunnybrook Research Institute, Toronto, Ontario, Canada
- Department of Ophthalmology and Vision Sciences, University of Toronto, Toronto, Ontario, Canada
- Department of Biochemistry, University of Toronto, Toronto, Ontario, Canada
| | - Alissa Pak
- Biological Sciences Platform, Sunnybrook Research Institute, Toronto, Ontario, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
| | - Luke Ajay David
- Biological Sciences Platform, Sunnybrook Research Institute, Toronto, Ontario, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
- Department of Ophthalmology and Vision Sciences, University of Toronto, Toronto, Ontario, Canada
| | - Edwin van Oosten
- Biological Sciences Platform, Sunnybrook Research Institute, Toronto, Ontario, Canada
- Department of Biochemistry, University of Toronto, Toronto, Ontario, Canada
| | - Rajiv Dixit
- Biological Sciences Platform, Sunnybrook Research Institute, Toronto, Ontario, Canada
- Department of Biochemistry, University of Toronto, Toronto, Ontario, Canada
| | - Laura M. Vecchio
- Biological Sciences Platform, Sunnybrook Research Institute, Toronto, Ontario, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
| | - Dhruv Nimesh Mehta
- Biological Sciences Platform, Sunnybrook Research Institute, Toronto, Ontario, Canada
| | - Ren Minamisono
- Biological Sciences Platform, Sunnybrook Research Institute, Toronto, Ontario, Canada
| | - Isabelle Aubert
- Biological Sciences Platform, Sunnybrook Research Institute, Toronto, Ontario, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
| | - Carol Schuurmans
- Biological Sciences Platform, Sunnybrook Research Institute, Toronto, Ontario, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
- Department of Ophthalmology and Vision Sciences, University of Toronto, Toronto, Ontario, Canada
- Department of Biochemistry, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
3
|
Pérez-Fernández V, Thananjeyan AL, Ullah F, Münch G, Cameron M, Gyengesi E. The effects of a highly bioavailable curcumin Phytosome TM preparation on the retinal architecture and glial reactivity in the GFAP-IL6 mice. FRONTIERS IN OPHTHALMOLOGY 2023; 3:1205542. [PMID: 38983084 PMCID: PMC11182199 DOI: 10.3389/fopht.2023.1205542] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Accepted: 09/08/2023] [Indexed: 07/11/2024]
Abstract
Uncontrolled, chronic inflammation in the retina can disturb retinal structure and function leading to impaired visual function. For the first time, in a mouse model of chronic neuroinflammation (GFAP-IL6), we investigated the impact of chronic glial activation on the retinal microglia population and structure. In addition, we tested a curcumin PhytosomeTM preparation with enhanced bioavailability to investigate the effects of a cytokine-suppressing anti-inflammatory drug on retinal architecture. Curcumin PhytosomeTM was fed to 3-month old GFAP-IL6 mice for 4 weeks and compared to their untreated GFAP-IL6 counterparts as well as wild type mice on control diet. Microglial numbers and morphology together with neuronal numbers were characterized using immunohistochemistry and cell reconstruction in the retina, using retinal wholemount and slices. GFAP-IL6 mice showed a significant increase in Iba1-labelled mononuclear phagocytes, including microglia, and displayed altered glial morphology. This resulted in a reduction in cone density and a thinning of the retinal layers compared to wild type mice. Curcumin PhytosomeTM treatment contributed to decreased microglial density, significantly decreasing both soma and cell size compared to control diet, as well as preventing the thinning of the retinal layers. This study is the first to characterize the impact of chronic retinal inflammation in the GFAP-IL6 mouse and the therapeutic benefit of enhanced bioavailable curcumin PhytosomeTM to significantly reduce microglia density and prevent neuronal loss. These data suggest that curcumin could be used as a complementary therapy alongside traditional treatments to reduce associated retinal inflammation in a variety of retinal diseases.
Collapse
Affiliation(s)
- Víctor Pérez-Fernández
- Department of Anatomy and Cell Biology, Western Sydney University, Campbelltown, NSW, Australia
| | | | - Faheem Ullah
- Department of Pharmacology, Western Sydney University, Campbelltown, NSW, Australia
- Neurosurgery, Robert Wood Johnson Medical School, Rutgers University, Piscataway, NJ, United States
| | - Gerald Münch
- Department of Pharmacology, Western Sydney University, Campbelltown, NSW, Australia
| | - Morven Cameron
- Department of Anatomy and Cell Biology, Western Sydney University, Campbelltown, NSW, Australia
| | - Erika Gyengesi
- Department of Pharmacology, Western Sydney University, Campbelltown, NSW, Australia
| |
Collapse
|
4
|
Saad El-Din S, Ahmed Rashed L, Saeed Mohamed D, Eissa M, Mohammad Raafat Hamed R, Elsayed Hussein R. Regulatory Role of circRNA-0067835 in Behcet Disease through Targeting Micro RNA-155: Implication of ATG1, AKT and MTOR. Rep Biochem Mol Biol 2023; 12:195-204. [PMID: 37724157 PMCID: PMC10505462 DOI: 10.52547/rbmb.12.1.195] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Accepted: 05/30/2023] [Indexed: 09/20/2023]
Abstract
Background Autophagy has been proven to contribute to maintaining eukaryotic cells' normal intracellular homeostasis, whereas autophagy malfunction may predispose to Behcet Disease (BD). The accumulation of the products of autophagic degradation as well as impairment in autophagic flux in cases with BD, may be attributed to dysregulated miRNA-155 expression. This study attempts to determine the contribution of circRNA-0067835 in miRNA-155-mediated modulation of the autophagy axis as well as to investigate its impact on the production of pro-inflammatory cytokines in BD. Methods This study was carried out on 40 cases with BD and 40 healthy control subjects. The collection of serum samples was done before performing a real-time PCR to estimate the relative gene expression of ATG1, AKT, miRNA-155, mTOR, TAB2, and circRNA-0067835. Additionally, IL-1β, IL-17, and TNF-α serum levels were determined by ELISA. Results Behcet Disease (BD) patients had significantly upregulated circRNA-0067835, with subsequent downregulation of its target gene, miRNA-155 than controls (P<0.05). In addition, decreased miRNA-155 gene expression was correlated with significantly increased TAB2 gene expression levels in BD patients compared to the controls (P<0.05). Furthermore, enhanced production of pro-inflammatory cytokines was detected in cases with BD than in controls. Conclusion The correlation between circRNA-0067835 and miRNA-155 fairly contributes to the regulation of cytokine production in BD via the modulation of autophagy. The investigation of the circRNA-0067835 and the microRNA-155 and their downstream adaptor molecules could be a potential therapeutic agent for BD.
Collapse
Affiliation(s)
- Shimaa Saad El-Din
- Medical Biochemistry and Molecular Biology Department. Faculty of Medicine, Cairo University, Egypt.
| | - Laila Ahmed Rashed
- Medical Biochemistry and Molecular Biology Department. Faculty of Medicine, Cairo University, Egypt.
| | - Doaa Saeed Mohamed
- Medical Biochemistry and Molecular Biology Department. Faculty of Medicine, Cairo University, Egypt.
| | - Mervat Eissa
- Rheumatology and Rehabilitation Department. Faculty of Medicine, Cairo University, Egypt.
| | | | - Rania Elsayed Hussein
- Medical Biochemistry and Molecular Biology Department. Faculty of Medicine, Cairo University, Egypt.
| |
Collapse
|
5
|
Sirolimus loaded chitosan functionalized PLGA nanoparticles protect against sodium iodate-induced retinal degeneration. J Drug Deliv Sci Technol 2023. [DOI: 10.1016/j.jddst.2023.104369] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/18/2023]
|
6
|
Protective effects of Panax ginseng berry extract on blue light-induced retinal damage in ARPE-19 cells and mouse retina. J Ginseng Res 2023; 47:65-73. [PMID: 36644394 PMCID: PMC9834005 DOI: 10.1016/j.jgr.2022.04.002] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Revised: 03/30/2022] [Accepted: 04/11/2022] [Indexed: 01/18/2023] Open
Abstract
Background Age-related macular degeneration (AMD) is a significant visual disease that induces impaired vision and irreversible blindness in the elderly. However, the effects of ginseng berry extract (GBE) on the retina have not been studied. Therefore, this study aimed to investigate the protective effects of GBE on blue light (BL)-induced retinal damage and elucidate its underlying mechanisms in human retinal pigment epithelial cells (ARPE-19 cells) and Balb/c retina. Methods To investigate the effects and underlying mechanisms of GBE on retinal damage in vitro, we performed cell viability assay, pre-and post-treatment of sample, reactive oxygen species (ROS) assay, quantitative real-time PCR (qRT-PCR), and western immunoblotting using A2E-laden ARPE-19 cells with BL exposure. In addition, Balb/c mice were irradiated with BL to induce retinal degeneration and orally administrated with GBE (50, 100, 200 mg/kg). Using the harvested retina, we performed histological analysis (thickness of retinal layers), qRT-PCR, and western immunoblotting to elucidate the effects and mechanisms of GBE against retinal damage in vivo. Results GBE significantly inhibited BL-induced cell damage in ARPE-19 cells by activating the SIRT1/PGC-1α pathway, regulating NF-kB translocation, caspase 3 activation, PARP cleavage, expressions of apoptosis-related factors (BAX/BCL-2, LC3-Ⅱ, and p62), and ROS production. Furthermore, GBE prevented BL-induced retinal degeneration by restoring the thickness of retinal layers and suppressed inflammation and apoptosis via regulation of NF-kB and SIRT1/PGC-1α pathway, cleavage of caspase 3 and PARP, and expressions of apoptosis-related factors in vivo. Conclusions GBE could be a potential agent to prevent dry AMD and progression to wet AMD.
Collapse
|
7
|
Alotaibi M, Al-Aqil F, Alqahtani F, Alanazi M, Nadeem A, Ahmad SF, Lapresa R, Alharbi M, Alshammari A, Alotaibi M, Saleh T, Alrowis R. Alleviation of cisplatin-induced neuropathic pain, neuronal apoptosis, and systemic inflammation in mice by rapamycin. Front Aging Neurosci 2022; 14:891593. [PMID: 36248001 PMCID: PMC9554141 DOI: 10.3389/fnagi.2022.891593] [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: 03/07/2022] [Accepted: 08/15/2022] [Indexed: 11/30/2022] Open
Abstract
Platinum-based chemotherapeutic treatment of cancer patients is associated with debilitating adverse effects. Several adverse effects have been well investigated, and can be managed satisfactorily, but chemotherapy-induced peripheral neuropathy (CIPN) remains poorly treated. Our primary aim in this study was to investigate the neuroprotective effect of the immunomodulatory drug rapamycin in the mitigation of cisplatin-induced neurotoxicity. Pain assays were performed in vivo to determine whether rapamycin would prevent or significantly decrease cisplatin-induced neurotoxicity in adult male Balb/c mice. Neuropathic pain induced by both chronic and acute exposure to cisplatin was measured by hot plate assay, cold plate assay, tail-flick test, and plantar test. Rapamycin co-treatment resulted in significant reduction in cisplatin-induced nociceptive-like symptoms. To understand the underlying mechanisms behind rapamycin-mediated neuroprotection, we investigated its effect on certain inflammatory mediators implicated in the propagation of chemotherapy-induced neurotoxicity. Interestingly, cisplatin was found to significantly increase peripheral IL-17A expression and CD8- T cells, which were remarkably reversed by the pre-treatment of mice with rapamycin. In addition, rapamycin reduced the cisplatin-induced neuronal apoptosis marked by decreased neuronal caspase-3 activity. The rapamycin neuroprotective effect was also associated with reversal of the changes in protein expression of p21Cip1, p53, and PUMA. Collectively, rapamycin alleviated some features of cisplatin-induced neurotoxicity in mice and can be further investigated for the treatment of cisplatin-induced peripheral neuropathy.
Collapse
Affiliation(s)
- Moureq Alotaibi
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
- *Correspondence: Moureq Alotaibi,
| | - Faten Al-Aqil
- Deanship of Scientific Research, King Saud University, Riyadh, Saudi Arabia
| | - Faleh Alqahtani
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Miteb Alanazi
- Pharmacy Services, King Saud University Medical City, Riyadh, Saudi Arabia
| | - Ahmed Nadeem
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Sheikh F. Ahmad
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Rebeca Lapresa
- Institute of Functional Biology and Genomics, Consejo Superior de Investigaciones Científicas (CSIC), University of Salamanca, Salamanca, Spain
- Institute of Biomedical Research of Salamanca, University Hospital of Salamanca, Consejo Superior de Investigaciones Científicas (CSIC), University of Salamanca, Salamanca, Spain
| | - Metab Alharbi
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Abdulrahman Alshammari
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Muteb Alotaibi
- Department of Neurology, Prince Sultan Military Medical City, Riyadh, Saudi Arabia
| | - Tareq Saleh
- Department of Basic Medical Sciences, Faculty of Medicine, The Hashemite University, Zarqa, Jordan
| | - Raed Alrowis
- Department of Periodotics and Community Dentistry, College of Dentistry, King Saud University, Riyadh, Saudi Arabia
| |
Collapse
|
8
|
Zaini LM, Kartasasmita AS, Gondhowiardjo TD, Syukri M, Lesmana R. Potential molecular mechanism of action of sodium-glucose co-transporter 2 inhibitors in the prevention and management of diabetic retinopathy. EXPERT REVIEW OF OPHTHALMOLOGY 2022. [DOI: 10.1080/17469899.2022.2111302] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/15/2022]
Affiliation(s)
- Lia Meuthia Zaini
- Faculty of Medicine, Padjadjaran University, Bandung, Indonesia
- Faculty of Medicine, Syiah Kuala University, Banda Aceh, Indonesia
- Department of Medicine, Zainoel Abidin Hospital, Banda Aceh
| | - Arief S Kartasasmita
- Faculty of Medicine, Padjadjaran University, Bandung, Indonesia
- Department of Ophthalmology, Cicendo Eye Hospital, Bandung, Indonesia
| | - Tjahjono D Gondhowiardjo
- Faculty of Medicine, Indonesia University, Jakarta, Indonesia
- Department of Ophthalmology, Cipto Mangunkusumo Hospital, Jakarta, Indonesia
| | - Maimun Syukri
- Faculty of Medicine, Syiah Kuala University, Banda Aceh, Indonesia
| | - Ronny Lesmana
- Faculty of Medicine, Padjadjaran University, Bandung, Indonesia
| |
Collapse
|
9
|
Mei L, Yu M, Liu Y, Weh E, Pawar M, Li L, Besirli CG, Schwendeman AA. Synthetic high-density lipoprotein nanoparticles delivering rapamycin for the treatment of age-related macular degeneration. NANOMEDICINE : NANOTECHNOLOGY, BIOLOGY, AND MEDICINE 2022; 44:102571. [PMID: 35623563 PMCID: PMC10655893 DOI: 10.1016/j.nano.2022.102571] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Revised: 04/25/2022] [Accepted: 05/18/2022] [Indexed: 05/03/2023]
Abstract
Synthetic high-density lipoprotein (sHDL) and rapamycin (Rap) have both been shown to be potential treatments for age-related macular degeneration (AMD). The low aqueous solubility of Rap, however, limits its therapeutic utility. Here we used an Apolipoprotein A-I mimetic peptide and phospholipid-based sHDL for the intravitreal delivery of Rap. By incorporation of Rap in sHDL nanoparticles (sHDL-Rap), we achieve 125-fold increase in drug aqueous concentration. When applied in vitro to retinal pigment epithelium cells, sHDL-Rap exhibited the abilities to efflux cholesterol, neutralize endotoxin, and suppress NF-κB activation. As an mTOR inhibitor, Rap induced autophagy and inhibited NF-κB-mediated pro-inflammatory signaling. Additionally, a greater reduction in lipofuscin accumulation and increased anti-inflammatory effects were achieved by sHDL-Rap relative to free drug or sHDL alone. In vivo studies demonstrated that sHDL reached the target retina pigment epithelium (RPE) layer following intravitreal administration in rats. These results suggest that sHDL-Rap holds potential as a treatment for AMD.
Collapse
Affiliation(s)
- Ling Mei
- Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, MI 48109, United States; Engineering Research Center for Pharmaceuticals and Equipments of Sichuan Province, Sichuan Industrial Institute of Antibiotics, School of Pharmacy, Chengdu University, Chengdu 610106, China
| | - Minzhi Yu
- Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, MI 48109, United States
| | - Yayuan Liu
- Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, MI 48109, United States
| | - Eric Weh
- Kellogg Eye Center, Department of Ophthalmology and Visual Sciences, University of Michigan, Ann Arbor, MI 48109, United States
| | - Mercy Pawar
- Kellogg Eye Center, Department of Ophthalmology and Visual Sciences, University of Michigan, Ann Arbor, MI 48109, United States
| | - Li Li
- Department of Pharmaceutical Sciences, Shenyang Pharmaceutical University, China
| | - Cagri G Besirli
- Kellogg Eye Center, Department of Ophthalmology and Visual Sciences, University of Michigan, Ann Arbor, MI 48109, United States
| | - Anna A Schwendeman
- Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, MI 48109, United States; Biointerfaces Institute, University of Michigan, Ann Arbor, MI 48109, United States.
| |
Collapse
|
10
|
Beaudry AG, Law ML. Leucine Supplementation in Cancer Cachexia: Mechanisms and a Review of the Pre-Clinical Literature. Nutrients 2022; 14:nu14142824. [PMID: 35889781 PMCID: PMC9323748 DOI: 10.3390/nu14142824] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Revised: 07/06/2022] [Accepted: 07/06/2022] [Indexed: 12/16/2022] Open
Abstract
Cancer cachexia (CC) is a complex syndrome of bodily wasting and progressive functional decline. Unlike starvation, cachexia cannot be reversed by increased energy intake alone. Nonetheless, targeted nutritional support is a necessary component in multimodal syndrome management. Due to the highly catabolic nature of cancer cachexia, amino acid supplementation has been proposed. Interestingly, leucine has been found to increase protein synthesis and decrease protein degradation via mTORC1 pathway activation. Multiple pre-clinical studies have explored the impact of leucine supplementation in cachectic tumor-bearing hosts. Here, we provide an overview of leucine’s proposed modes of action to preserve lean mass in cachexia and review the current pre-clinical literature related to leucine supplementation during CC. Current research indicates that a leucine-rich diet may attenuate CC symptomology; however, these works are difficult to compare due to methodological differences. There is need for further pre-clinical work exploring leucine’s potential ability to modulate protein turnover and immune response during CC, as well as the impact of additive leucine on tumor growth.
Collapse
Affiliation(s)
- Anna G. Beaudry
- Department of Health, Human Performance, and Recreation, Robbins College of Health and Human Sciences, Baylor University, Waco, TX 76706, USA
- Correspondence:
| | - Michelle L. Law
- Department of Human Sciences and Design, Robbins College of Health and Human Sciences, Baylor University, Waco, TX 76706, USA;
| |
Collapse
|
11
|
Park K, Lee MS. Current Status of Autophagy Enhancers in Metabolic Disorders and Other Diseases. Front Cell Dev Biol 2022; 10:811701. [PMID: 35237600 PMCID: PMC8882819 DOI: 10.3389/fcell.2022.811701] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Accepted: 01/13/2022] [Indexed: 12/21/2022] Open
Abstract
Autophagy is pivotal in the maintenance of organelle function and intracellular nutrient balance. Besides the role of autophagy in the homeostasis and physiology of the individual tissues and whole organism in vivo, dysregulated autophagy has been incriminated in the pathogenesis of a variety of diseases including metabolic diseases, neurodegenerative diseases, cardiovascular diseases, inflammatory or immunological disorders, cancer and aging. Search for autophagy modulators has been widely conducted to amend dysregulation of autophagy or pharmacologically modulate autophagy in those diseases. Current data support the view that autophagy modulation could be a new modality for treatment of metabolic syndrome associated with lipid overload, human-type diabetes characterized by deposition of islet amyloid or other diseases including neurodegenerative diseases, infection and cardiovascular diseases. While clinically available bona fide autophagy modulators have not been developed yet, it is expected that on-going investigation will lead to the development of authentic autophagy modulators that can be safely administered to patients in the near future and will open a new horizon for treatment of incurable or difficult diseases.
Collapse
|
12
|
Xie D, Zhao T, Zhang X, Kui L, Wang Q, Wu Y, Zheng T, Ma P, Zhang Y, Molteni H, Geng R, Yang Y, Li B, Zheng QY. Autophagy Contributes to the Rapamycin-Induced Improvement of Otitis Media. Front Cell Neurosci 2022; 15:753369. [PMID: 35153674 PMCID: PMC8832103 DOI: 10.3389/fncel.2021.753369] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Accepted: 12/31/2021] [Indexed: 12/02/2022] Open
Abstract
Otitis media (OM) is a pervasive disease that involves hearing loss and severe complications. In our previous study, we successfully established a mouse model of human OM using Tlr2tm1Kir (TLR2-/-) mice with middle ear (ME) inoculation of streptococcal peptidoglycan-polysaccharide (PGPS). In this study, we found that hearing loss and OM infections in OM mice were significantly alleviated after treatment with rapamycin (RPM), a widely used mechanistic target of RPM complex 1 (mTORC1) inhibitor and autophagy inducer. First of all, we tested the activity of mTORC1 by evaluating p-S6, Raptor, and mTOR protein expression. The data suggested that the protein expression level of p-S6, Raptor and mTOR are decreased in TLR2-/- mice after the injection of PGPS. Furthermore, our data showed that both the autophagosome protein LC3-II, Beclin-1, ATG7, and autophagy substrate protein p62 accumulated at higher levels in mice with OM than in OM-negative mice. The expression of lysosomal-associated proteins LAMP1, Cathepsin B, and Cathepsin D increased in the OM mice compared with OM-negative mice. Rab7 and Syntaxin 17, which is necessary for the fusion of autophagosomes with lysosomes, are reduced in the OM mice. In addition, data also described that the protein expression level of p-S6, mTOR and Raptor are lower than PGPS group after RPM treatment. The accumulation of LC3-II, Beclin-1, and ATG7 are decreased, and the expression of Rab7 and Syntaxin 17 are increased significantly after RPM treatment. Our results suggest that autophagy impairment is involved in PGPS-induced OM and that RPM improves OM at least partly by relieving autophagy impairment. Modulating autophagic activity by RPM may be a possible effective treatment strategy for OM.
Collapse
Affiliation(s)
- Daoli Xie
- Hearing and Speech Rehabilitation Institute, College of Special Education, Binzhou Medical University, Yantai, China
| | - Tong Zhao
- Hearing and Speech Rehabilitation Institute, College of Special Education, Binzhou Medical University, Yantai, China
| | - Xiaolin Zhang
- Department of Otolaryngology-Head and Neck Surgery, Binzhou Medical University Hospital, Binzhou, China
| | - Lihong Kui
- Hearing and Speech Rehabilitation Institute, College of Special Education, Binzhou Medical University, Yantai, China
| | - Qin Wang
- Hearing and Speech Rehabilitation Institute, College of Special Education, Binzhou Medical University, Yantai, China
| | - Yuancheng Wu
- Hearing and Speech Rehabilitation Institute, College of Special Education, Binzhou Medical University, Yantai, China
| | - Tihua Zheng
- Hearing and Speech Rehabilitation Institute, College of Special Education, Binzhou Medical University, Yantai, China
| | - Peng Ma
- Department of Genetics, School of Pharmacy, Binzhou Medical University, Yantai, China
| | - Yan Zhang
- Department of Otolaryngology, Head and Neck Surgery, Second Affiliated Hospital, Xi’an Jiaotong University School of Medicine, Xi’an, China
| | - Helen Molteni
- Department of Otolaryngology, Head and Neck Surgery, Case Western Reserve University, Cleveland, OH, United States
| | - Ruishuang Geng
- Hearing and Speech Rehabilitation Institute, College of Special Education, Binzhou Medical University, Yantai, China
| | - Ying Yang
- Hearing and Speech Rehabilitation Institute, College of Special Education, Binzhou Medical University, Yantai, China
| | - Bo Li
- Hearing and Speech Rehabilitation Institute, College of Special Education, Binzhou Medical University, Yantai, China
| | - Qing Yin Zheng
- Department of Otolaryngology, Head and Neck Surgery, Case Western Reserve University, Cleveland, OH, United States
| |
Collapse
|
13
|
Ma J, Zhang Y, Sugai T, Kubota T, Keino H, El-Salhy M, Ozaki M, Umezawa K. Inhibition of Cellular and Animal Inflammatory Disease Models by NF-κB Inhibitor DHMEQ. Cells 2021; 10:2271. [PMID: 34571920 PMCID: PMC8466912 DOI: 10.3390/cells10092271] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2021] [Revised: 08/27/2021] [Accepted: 08/31/2021] [Indexed: 12/26/2022] Open
Abstract
General inflammatory diseases include skin inflammation, rheumatoid arthritis, inflammatory bowel diseases, sepsis, arteriosclerosis, and asthma. Although these diseases have been extensively studied, most of them are still difficult to treat. Meanwhile, NF-κB is a transcription factor promoting the expression of many inflammatory mediators. NF-κB is likely to be involved in the mechanism of most inflammatory diseases. We discovered a specific NF-κB inhibitor, dehydroxymethylepoxyquinomicin (DHMEQ), about 20 years ago by molecular design from a natural product. It directly binds to and inactivates NF-κB components. It has been widely used to suppress cellular and animal inflammatory disease models and was shown to be potent in vivo anti-inflammatory activity without any toxicity. We have prepared ointment of DHMEQ for the treatment of severe skin inflammation. It inhibited inflammatory cytokine expressions and lowered the clinical score in mouse models of atopic dermatitis. Intraperitoneal (IP) administration of DHMEQ ameliorated various disease models of inflammation, such as rheumatoid arthritis, sepsis, and also graft rejection. It has been suggested that inflammatory cells in the peritoneal cavity would be important for most peripheral inflammation. In the present review, we describe the synthesis, mechanism of action, and cellular and in vivo anti-inflammatory activities and discuss the clinical use of DHMEQ for inflammatory diseases.
Collapse
Affiliation(s)
- Jun Ma
- Shenzhen Wanhe Pharmaceutical Co., Ltd., Shenzhen 518107, China;
| | - Yuyang Zhang
- School of Life Science and Biopharmaceutics, Shenyang Pharmaceutical University, Shenyang 110016, China;
| | - Takeshi Sugai
- Faculty of Pharmacy, Keio University, Tokyo 105-8512, Japan;
| | - Tetsuo Kubota
- Department of Medical Technology, Tsukuba International University, Tsuchiura 300-0051, Japan;
| | - Hiroshi Keino
- Department of Ophthalmology, Kyorin University School of Medicine, Tokyo 181-8611, Japan;
| | - Magdy El-Salhy
- Department of Medicine, Stord Helse-Fonna Hospital, Tysevegen 64, 54 16 Stord, Norway;
| | - Michitaka Ozaki
- Department of Biological Response and Regulation, Faculty of Health Sciences, Hokkaido University, Sapporo 060-0812, Japan;
| | - Kazuo Umezawa
- Department of Molecular Target Medicine, Aichi Medical University, Nagakute 480-1195, Japan
| |
Collapse
|
14
|
Niu Z, Shi Y, Li J, Qiao S, Du S, Chen L, Tian H, Wei L, Cao H, Wang J, Gao L. Protective effect of rapamycin in models of retinal degeneration. Exp Eye Res 2021; 210:108700. [PMID: 34245755 DOI: 10.1016/j.exer.2021.108700] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Revised: 06/25/2021] [Accepted: 07/06/2021] [Indexed: 12/29/2022]
Abstract
Age-related macular degeneration (AMD) is a complex retinal disease with no viable treatment strategy. The causative mechanistic pathway for this disease is not yet clear. Therefore, it is highly warranted to screen effective drugs to treat AMD. Rapamycin are known to inhibit inflammation and has been widely used in the clinic as an immunosuppressant. This study aimed to investigate the protective effect of rapamycin on the AMD retinal degeneration model. The AMD models were established by injection of 35 mg/kg sodium iodate (NaIO3) into the tail vein. Then the treated mice intraperitoneally received rapamycin (2 mg/kg) once a day. The histomorphological analysis showed that rapamycin could inhibit retinal structure damage and apoptosis. Experiments revealed that rapamycin significantly attenuated inflammatory response and oxidative stress. Our experimental results demonstrated that rapamycin has protected the retinal against degeneration induced by NaIO3. The therapeutic effect was more significant after 7 days of treatment. Therefore, our study potentially provides a powerful experimental support for the treatment of AMD.
Collapse
Affiliation(s)
- Zhanyu Niu
- School of Life Sciences, Lanzhou University, Lanzhou, 730000, China.
| | - Yongpeng Shi
- School of Life Sciences, Lanzhou University, Lanzhou, 730000, China.
| | - Jiande Li
- School of Life Sciences, Lanzhou University, Lanzhou, 730000, China.
| | - Shufan Qiao
- Colloge of Life Sciences, Northwest Normal University, Lanzhou, 730000, China.
| | - Shaobo Du
- School of Stomatology of Lanzhou University, Lanzhou, 730000, China.
| | - Linchi Chen
- School of Life Sciences, Lanzhou University, Lanzhou, 730000, China.
| | - Huanbing Tian
- School of Life Sciences, Lanzhou University, Lanzhou, 730000, China.
| | - Li Wei
- School of Life Sciences, Lanzhou University, Lanzhou, 730000, China.
| | - Hanwen Cao
- School of Life Sciences, Lanzhou University, Lanzhou, 730000, China.
| | - Ji Wang
- School of Life Sciences, Lanzhou University, Lanzhou, 730000, China.
| | - Lan Gao
- School of Life Sciences, Lanzhou University, Lanzhou, 730000, China.
| |
Collapse
|
15
|
Plasma Rich in Growth Factors Promotes Autophagy in ARPE19 Cells in Response to Oxidative Stress Induced by Blue Light. Biomolecules 2021; 11:biom11070954. [PMID: 34203504 PMCID: PMC8301887 DOI: 10.3390/biom11070954] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Revised: 06/23/2021] [Accepted: 06/24/2021] [Indexed: 12/15/2022] Open
Abstract
Age-related macular degeneration (AMD) causes the degeneration of photoreceptors and retinal cells leading to vision loss in older subjects. Among possible exogenous risk factors, it has been recently proposed that long-term exposure to blue light could aggravate the course of AMD. In the search for therapeutic options, plasma rich in growth factors (PRGF) has been shown to enhance cell antioxidant pathways and protect photoreceptors against the harm produced by blue light, although its mechanism of action remains unknown. One possible mechanism, autophagy, is one of the most conservative cell renewal systems used in eukaryotes to destroy cellular components that have been damaged by some kind of insult. The oxidative stress of exposure to blue light is known to induce cell autophagy. In this study, we examined the combined effects on autophagy of blue light and PRGF in a retinal cell line, ARPE19. In response to treatment with both PRGF and blue light, we detected the modulated expression of autophagy markers such as NF-kB, p62/sqstm1, Atg5, LC3 and Beclin1, and inflammatory markers such as IL1B and IL18. Our findings suggest that PRGF promotes cell autophagy in response to exposure to blue light.
Collapse
|
16
|
Liang L, Zhou Q, Feng L. Decreased microRNA-155 in Behcet's disease leads to defective control of autophagy thereby stimulating excessive proinflammatory cytokine production. Arthritis Res Ther 2021; 23:135. [PMID: 33957967 PMCID: PMC8101176 DOI: 10.1186/s13075-021-02517-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2020] [Accepted: 04/20/2021] [Indexed: 01/09/2023] Open
Abstract
BACKGROUND Earlier, we reported that the microRNA (miR)-155 expression in dendritic cells (DCs) from Behcet's disease (BD) patients was decreased and affected cytokine production of DCs. In this study, we investigated the mechanisms whereby miR-155 regulates cytokine production by DCs. METHODS The formation of autophagosomes in DCs was detected by transmission electron microscopy. Western blotting was used to detect the protein levels of LC3, Beclin-1, P62, p-mTOR, and p-Akt in DCs. TNF-α, IL-6, and IL-1β expression were investigated by ELISA. MiR-155 mimics were transfected to DCs to evaluate its effects on autophagy and cytokine production. RNA interference was used to downregulate the expression of TAB2. RESULTS The formation of autophagosomes was found in DCs of active BD patients. The expressions of LC3-II, Beclin-1, and P62 were significantly increased in DCs of active BD patients compared to that of inactive BD patients and healthy controls. The expressions of IL-6, IL-1β, and TNF-α were significantly increased in DCs of active BD patients compared to that of healthy controls. The autophagy promoter (3-MA) and inhibitor (rapamycin) significantly decreased or increased the expression of TNF-α, IL-6, and IL-1β by DCs. The expression of LC3-II and Beclin-1 was significantly increased, but the expression of P62 proteins was decreased in DCs transfected with miR-155 mimics or after TAB2 was downregulated. The expression of TNF-α, IL-6, and IL-1β was decreased in DCs after miR-155 was upregulated or TAB2 was downregulated. The ratios of p-Akt/Akt and p-mTOR/mTOR were decreased in DCs after miR-155 was upregulated. CONCLUSIONS These results suggest that miR-155 affects the production of TNF-α, IL-6, and IL-1β by DCs through activation of the Akt/mTOR signaling pathway and by affecting the process of autophagy.
Collapse
Affiliation(s)
- Liang Liang
- The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Lab of Ophthalmology, Chongqing Eye Institute, Chongqing Branch of National Clinical Research Center for Ocular Diseases, Chongqing, P. R. China.
| | - Qingyun Zhou
- The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Lab of Ophthalmology, Chongqing Eye Institute, Chongqing Branch of National Clinical Research Center for Ocular Diseases, Chongqing, P. R. China
| | - Lujia Feng
- The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Lab of Ophthalmology, Chongqing Eye Institute, Chongqing Branch of National Clinical Research Center for Ocular Diseases, Chongqing, P. R. China
| |
Collapse
|
17
|
Li HG, Tian WH, Qin CL, Ye RR, Liu DH, Liu HW. Uhrf1 regulates H3K9me2 modification of mTOR to inhibit the effect of autophagy in myocardial ischemia-reperfusion injury. Aging (Albany NY) 2021; 13:9704-9718. [PMID: 33744855 PMCID: PMC8064229 DOI: 10.18632/aging.202722] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2020] [Accepted: 11/15/2020] [Indexed: 11/25/2022]
Abstract
The regulation of mTOR and the dimethylation of histone H3 on lysine 9 (H3K9me2) H3K9me2 by Uhrf1 and the mechanism of autophagy regulation in myocardial ischemia-reperfusion injury (MIRI) were studied in vivo and in vitro. An in vitro I/R injury model was established using the primary mouse cardiomyocytes treated with H2O2. Subsequent analysis by qRT-PCR, western blot, and immunofluorescence indicated that overexpression of Uhrf1 significantly inhibited apoptosis of the H2O2-treated cardiomyocytes, reduced expression of apoptosis factors caspase-3 and Bax, and increased expression of apoptosis inhibitory factor Bcl-2. Furthermore, Uhrf1 was found to increase cardiomyocyte proliferation and promote the expression of mTOR, while the four expression peaks of H3K9me2 on the mTOR gene were inhibited by overexpression of Uhrf1. The expression of autophagy factors LC3, Beclin-1, and p-mTOR in Uhrf1-overexpressed cardiomyocytes was dramatically increased, and P62 expression was dramatically decreased. When an H3K9me2 inhibitor was added to the Uhrf1-knockdown cardiomyocytes, the expression of mTOR was increased, the expression of LC3, Beclin-1, and p-mTOR was decreased, and P62 expression was significantly increased. In the present study, Uhrf1 exhibits a protective function in MIRI, reducing the apoptosis of cardiomyocytes while increasing their proliferation and viability.
Collapse
Affiliation(s)
- Han-Geng Li
- Department of Histology and Embryology, College of Basic Medicine, Harbin Medical University, Harbin 150081, China
| | - Wen-Hua Tian
- Emergency Department, The Fourth Hospital of Harbin Medical University, Harbin 150081, China
| | - Cun-Lan Qin
- Department of Histology and Embryology, College of Basic Medicine, Harbin Medical University, Harbin 150081, China
| | - Rong-Rong Ye
- Department of Histology and Embryology, College of Basic Medicine, Harbin Medical University, Harbin 150081, China
| | - Dong-Hua Liu
- Department of Histology and Embryology, College of Basic Medicine, Harbin Medical University, Harbin 150081, China
| | - Hui-Wen Liu
- Department of Histology and Embryology, College of Basic Medicine, Harbin Medical University, Harbin 150081, China
| |
Collapse
|
18
|
Salas-Ambrosio PJ, Bernad-Bernad MJ, Linares-Alba MA, García-Santisteban R, Tonix-Aburto LA, Ornelas-Lobato GJ, Gracia-Mora I, Rivera-Huerta M, Sánchez-Bartez F, Rico-Morales H, García-Sánchez GA. Toxicity Evaluation of a Novel Rapamycin Liposomal Formulation After Subconjunctival and Intravitreal Injection. J Ocul Pharmacol Ther 2021; 37:261-276. [PMID: 33691483 DOI: 10.1089/jop.2020.0108] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Purpose: Safety and toxicity evaluation of a novel, liposome-encapsulated rapamycin formulation, intended for autoimmune ocular disorders. Methods: The formulation was assessed by micronucleus polychromatic erythrocyte production, irritability by Hen's Egg Test-Chorioallantoic Membrane (HET CAM), sterility, and pyrogenicity testing. Subconjunctival (SCJ) and intravitreal (IVT) administration of the formulation were performed to evaluate subacute and acute toxicity, respectively. Differences between groups in biochemical and hematological parameters were evaluated by analysis of variance and t-tests. Numeric score was assigned to histopathological classification. Electroretinography (ERG) testing was also performed. Data were analyzed by a 1 way no parametric Kruskal-Wallis and the Mann-Whitney tests. Significance was considered when P < 0.05. Results: No significant toxicity directly related to the preparation was detected. Micronucleus count, mucous irritation score, and pyrogenicity results were negative. Pathology demonstrated no damage related to the formulation after SCJ injection. After IVT injection, only lens injury associated with technique was observed. Retinal function was also conserved in ERG. Conclusions: The preparation evaluated offers a good toxicity and safety profile when injected in a SCJ or IVT manner in an animal model. A clinical trial conducted in humans is highly warranted, as it could reveal an alternative immunosuppressive treatment for ophthalmological immune-mediated pathologies.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Isabel Gracia-Mora
- Facultad de Química, Universidad Nacional Autónoma de México, Coyoacán, Mexico City
| | | | | | - Héctor Rico-Morales
- Facultad de Química, Universidad Nacional Autónoma de México, Coyoacán, Mexico City
| | | |
Collapse
|
19
|
Kutsyr O, Sánchez-Sáez X, Martínez-Gil N, de Juan E, Lax P, Maneu V, Cuenca N. Gradual Increase in Environmental Light Intensity Induces Oxidative Stress and Inflammation and Accelerates Retinal Neurodegeneration. Invest Ophthalmol Vis Sci 2021; 61:1. [PMID: 32744596 PMCID: PMC7441298 DOI: 10.1167/iovs.61.10.1] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Purpose Retinitis pigmentosa (RP) is a blinding neurodegenerative disease of the retina that can be affected by many factors. The present study aimed to analyze the effect of different environmental light intensities in rd10 mice retina. Methods C57BL/6J and rd10 mice were bred and housed under three different environmental light intensities: scotopic (5 lux), mesopic (50 lux), and photopic (300 lux). Visual function was studied using electroretinography and optomotor testing. The structural and morphological integrity of the retinas was evaluated by optical coherence tomography imaging and immunohistochemistry. Additionally, inflammatory processes and oxidative stress markers were analyzed by flow cytometry and western blotting. Results When the environmental light intensity was higher, retinal function decreased in rd10 mice and was accompanied by light-dependent photoreceptor loss, followed by morphological alterations, and synaptic connectivity loss. Moreover, light-dependent retinal degeneration was accompanied by an increased number of inflammatory cells, which became more activated and phagocytic, and by an exacerbated reactive gliosis. Furthermore, light-dependent increment in oxidative stress markers in rd10 mice retina pointed to a possible mechanism for light-induced photoreceptor degeneration. Conclusions An increase in rd10 mice housing light intensity accelerates retinal degeneration, activating cell death, oxidative stress pathways, and inflammatory cells. Lighting intensity is a key factor in the progression of retinal degeneration, and standardized lighting conditions are advisable for proper analysis and interpretation of experimental results from RP animal models, and specifically from rd10 mice. Also, it can be hypothesized that light protection could be an option to slow down retinal degeneration in some cases of RP.
Collapse
|
20
|
Ischemic postconditioning reduced myocardial ischemia-reperfusion injury: The roles of melatonin and uncoupling protein 3. Anatol J Cardiol 2020; 23:19-27. [PMID: 31911566 PMCID: PMC7141427 DOI: 10.14744/anatoljcardiol.2019.72609] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Objective: Protective effects of ischemic postconditioning (PostC) decrease/disappear with age and chronic heart diseases. Similarly, low serum melatonin levels have been reported in the same risk groups. The aims of this study were to investigate the effects of melatonin on the protection of PostC in ischemia–reperfusion (I/R)-induced infarct size and roles of uncoupling protein (UCP) 3, irisin, and nuclear factor kappa B (NFkB) levels. Methods: Rats were pinealectomized (Px) or sham operated (non-Px) 2 months before the I/R studies. The left main coronary artery was occluded for 30 min followed by 120 min reperfusion. PostC was induced with three cycles of R/I (10 s each) after ischemia. Results: The infarct size was found to be significantly higher in Px rats (54.68±1.5%) than in the control group (35.1±2.5%). PostC and melatonin administrations to non-Px rats significantly reduced the infarct size. On the other hand, PostC did not create a significant effect in Px rats, but protection was provided when PostC was co-administrated with melatonin. While significant decreases were detected in the UCP3 levels, irisin and NFkB levels increased with I/R and Px. Treatment with PostC and melatonin in non-Px groups and their co-administration in Px groups were found to return all the genes close to normal levels. Conclusion: The physiological and pharmacological concentrations of melatonin may play a role in the protection of PostC. In cases when physiological melatonin is reduced, such as aging and heart diseases, this protection may decrease, and this effect may be restored by melatonin replacement. PostC and melatonin may regulate energy metabolism and inflammatory mediators and protect mitochondria by affecting the UCP3, irisin, and NFkB levels.
Collapse
|
21
|
Li C, Li C, Lin J, Zhao G, Xu Q, Jiang N, Wang Q, Peng X, Zhu G, Jiang J. The Role of Autophagy in the Innate Immune Response to Fungal Keratitis Caused by Aspergillus fumigatus Infection. Invest Ophthalmol Vis Sci 2020; 61:25. [PMID: 32084267 PMCID: PMC7326573 DOI: 10.1167/iovs.61.2.25] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Purpose To determine the role of autophagy in the innate immune response to fungal keratitis (FK) caused by Aspergillus fumigatus infection. Methods Corneal samples obtained from patients and mice with FK were visualized via transmission electron microscopy (TEM). Autophagy-related proteins LC3B-II, Beclin-1, LAMP-1, and p62 in A. fumigatus-infected corneas of C57BL/6 mice were tested by Western blot. After treatment with autophagy inhibitors 3-methyladenine (3-MA), chloroquine (CQ), or inducer rapamycin, autophagy-related proteins were detected by Western blot. Corneas were photographed with slit lamp microscopy and pathological changes were observed by hematoxylin and eosin staining. Polymorphonuclear neutrophilic leukocytes (PMNs) were assessed by immunofluorescent staining and observed under TEM. The levels of CXCL-1, IL-1β, HMGB1, IL-18, TNF-α, and IL-10 were tested by reverse transcription polymerase chain reaction and Western blot. The quantification of fungal loads was detected and photographed. Results The accumulation of autophagosomes in corneas of patients and mice with FK was observed with TEM. The expression of LC3B-II, Beclin-1, and LAMP-1 was elevated in corneas after fungal infection, whereas p62 was reduced. Treatment with 3-MA or CQ upregulated clinical scores, pathological changes, and the expression of CXCL-1, IL-1β, HMGB1, IL-18, and TNF-α except IL-10. The morphology of PMNs was changed and PMN recruitment was increased in mice corneas treated with 3-MA or CQ, whereas rapamycin reduced the inflammatory response to keratitis. These results were statistically significant. Conclusions A. fumigatus infection increases the expression of autophagy in corneas. Autophagy plays an anti-inflammatory role in the innate immune response to A. fumigatus keratitis.
Collapse
|
22
|
Kawashima H, Ozawa Y, Toda E, Homma K, Osada H, Narimatsu T, Nagai N, Tsubota K. Neuroprotective and vision-protective effect of preserving ATP levels by AMPK activator. FASEB J 2020; 34:5016-5026. [PMID: 32090372 DOI: 10.1096/fj.201902387rr] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2019] [Revised: 01/17/2020] [Accepted: 01/22/2020] [Indexed: 12/24/2022]
Abstract
Progression of blinding diseases, such as age-related macular degeneration, is accelerated by light exposure. However, no particular intervention is applied to the photostress. Here, we report neuroprotective effects of the adenosine monophosphate (AMP)-activated protein kinase (AMPK) activator, 5-Aminoimidazole-4-carboxamide ribonucleotide (AICAR), on light-induced visual function impairment, photoreceptor disorders and death in mice. Increase in retinal ATP levels in response to photostress was transient, because oxygen consumption rate (OCR) and cytochrome c oxidase (CcO) activity were reduced under photostress. However, AICAR treatment preserved OCR, CcO activity, and high levels of retinal ATP after light exposure. AMPK knockdown in the photoreceptor-derived cell line revealed that AMPK targeted CcO activity. Further, our data indicated that photostress reduced mitochondrial respiratory function and ATP levels, while AICAR treatment promoted neuronal survival and retained visual function, stabilizing ATP levels through preserved CcO activity. The current study has provided proof of concept for providing cells with sufficient energy to promote cell survival in the presence of cellular stress. This is in contrast to the previous reports which primarily investigated therapeutic approaches to suppress stress signals. Hence, stabilization of the ATP supply may serve as a novel therapeutic approach to support tissue survival under stress and prevent neurodegeneration.
Collapse
Affiliation(s)
- Hirohiko Kawashima
- Laboratory of Retinal Cell Biology, Keio University School of Medicine, Tokyo, Japan.,Department of Ophthalmology, Keio University School of Medicine, Tokyo, Japan
| | - Yoko Ozawa
- Laboratory of Retinal Cell Biology, Keio University School of Medicine, Tokyo, Japan.,Department of Ophthalmology, Keio University School of Medicine, Tokyo, Japan
| | - Eriko Toda
- Laboratory of Retinal Cell Biology, Keio University School of Medicine, Tokyo, Japan
| | - Kohei Homma
- Laboratory of Retinal Cell Biology, Keio University School of Medicine, Tokyo, Japan
| | - Hideto Osada
- Laboratory of Retinal Cell Biology, Keio University School of Medicine, Tokyo, Japan
| | - Toshio Narimatsu
- Laboratory of Retinal Cell Biology, Keio University School of Medicine, Tokyo, Japan.,Department of Ophthalmology, Keio University School of Medicine, Tokyo, Japan
| | - Norihiro Nagai
- Laboratory of Retinal Cell Biology, Keio University School of Medicine, Tokyo, Japan.,Department of Ophthalmology, Keio University School of Medicine, Tokyo, Japan
| | - Kazuo Tsubota
- Department of Ophthalmology, Keio University School of Medicine, Tokyo, Japan
| |
Collapse
|
23
|
Gao Y, You X, Liu Y, Gao F, Zhang Y, Yang J, Yang C. Induction of autophagy protects human dental pulp cells from lipopolysaccharide-induced pyroptotic cell death. Exp Ther Med 2020; 19:2202-2210. [PMID: 32104285 PMCID: PMC7027320 DOI: 10.3892/etm.2020.8475] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2019] [Accepted: 11/01/2019] [Indexed: 02/06/2023] Open
Abstract
The NOD-like receptor protein 3/caspase-1 inflammasome can be activated in human dental pulp tissue and fibroblasts; however, the underlying mechanisms are poorly understood. In the present study, lipopolysaccharide (LPS) was used to treat dental pulp cells to establish an inflammation model. Cell viability was examined by sulforhodamine B assay. Interleukin (IL)-1β, caspase-1, microtubule-associated protein-1 light chain 3-II/I and p62 were determined by western blotting and ELISA. The phosphorylation (p-) levels of NF-κB and NF-κB inhibitor (IκB)α protein were observed by western blotting. The results demonstrated that LPS induced pyroptotic cell death in cultured dental pulp cells, which was supported by the increased levels of IL-1β, IL-18 and caspase-1. Rapamycin and 3-methyladenine (3-MA) were used to activate and inhibit autophagy, and it was observed that LPS increased autophagy and rapamycin reduced LPS-induced dental pulp cell pyroptosis. However, 3-MA aggravated LPS-induced dental pulp cell pyroptosis. In addition, LPS inhibited the expression of IκBα, but increased the expression of p-NF-κB. Compared with the LPS group, 3-MA further inhibited the expression of IκBα but promoted the expression of p-NF-κB. However, rapamycin produced the opposite results to LPS. Under LPS treatment, the NF-κB pathway inhibitor BAY11-7082 further enhanced the inhibitory effects of rapamycin, but inhibited the promoting effects of 3-MA on the protein expression levels of IL-1β and caspase-1. The results of the present study demonstrated that there is an important crosstalk between autophagy, pyroptosis and the NF-κB pathway, and that the modulation of pyroptosis in dental pulp cells may be a promising strategy to pulpitis therapy.
Collapse
Affiliation(s)
- Yang Gao
- Jiangsu Key Laboratory of Oral Disease, The Affiliated Hospital of Stomatology, Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China.,Department of Oral and Maxillofacial Surgery, The Affiliated Hospital of Stomatology, Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China.,Department of Stomatology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215000, P.R. China
| | - Xinran You
- Department of Nuclear Medicine, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, Jiangsu 215002, P.R. China
| | - Yubo Liu
- Department of Orthopedics, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, Jiangsu 215002, P.R. China
| | - Fei Gao
- Department of Nuclear Medicine, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, Jiangsu 215002, P.R. China
| | - Yuan Zhang
- Department of Head and Neck Oncology, The Affiliated Jiangsu Cancer Hospital of Nanjing Medical University, Nanjing, Jiangsu 210009, P.R. China
| | - Jianrong Yang
- Jiangsu Key Laboratory of Oral Disease, The Affiliated Hospital of Stomatology, Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China.,Department of Oral and Maxillofacial Surgery, The Affiliated Hospital of Stomatology, Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| | - Chen Yang
- Department of Nuclear Medicine, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, Jiangsu 215002, P.R. China
| |
Collapse
|
24
|
Yuan H, Wu G, Zhai X, Lu B, Meng B, Chen J. Melatonin and Rapamycin Attenuate Isoflurane-Induced Cognitive Impairment Through Inhibition of Neuroinflammation by Suppressing the mTOR Signaling in the Hippocampus of Aged Mice. Front Aging Neurosci 2019; 11:314. [PMID: 31803045 PMCID: PMC6877689 DOI: 10.3389/fnagi.2019.00314] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2019] [Accepted: 10/31/2019] [Indexed: 12/26/2022] Open
Abstract
Melatonin exerts neuroprotective effects on isoflurane-induced cognitive impairment. However, the underlying mechanism has yet to be elucidated. The present study sought to determine if melatonin confers its beneficial effects by acting on mammalian target of rapamycin (mTOR) and attenuates the neuroinflammation in the hippocampus of aged mice. A total of 72 male C57BL/6 mice, 16-month-old, were randomly and equally divided into six groups: (1) the control group (CON); (2) the rapamycin group (RAP); (3) the melatonin group (MEL); (4) the isoflurane group (ISO); (5) the rapamycin + isoflurane group (RAP + ISO); and (6) the melatonin + isoflurane group (MEL + ISO). RAP, RAP + ISO, MEL, MEL + ISO groups received 1 mg/kg/day mTOR inhibitor rapamycin solution or 10 mg/kg/day melatonin solution, respectively, intraperitoneally at 5:00 p.m. for 14 days consecutively. Mice in the CON and ISO groups were administered an equivalent volume of saline. Subsequently, ISO, RAP + ISO, and MEL + ISO groups were exposed to inhale 2% isoflurane for 4 h; the CON, RAP, and MEL mice received only the vehicle gas. Then, the memory function and spatial learning of the mice were examined via the Morris water maze (MWM) test. mTOR expression was detected via Western blot, whereas the concentration of inflammatory cytokines tumor necrosis factor (TNF)-α, interleukin (IL)-1β, IL-6 and that of melatonin was quantified with enzyme-linked immunosorbent assay (ELISA). Melatonin and rapamycin significantly ameliorated the isoflurane-induced cognitive impairment and also led to a decrease in the melatonin levels as well as the expression levels of TNF-α, IL-1β, IL-6, and p-mTOR in the hippocampus. In conclusion, these results showed that melatonin and rapamycin attenuates mTOR expression while affecting the downstream proinflammatory cytokines. Thus, these molecular findings could be associated with an improved cognitive function in mice exposed to isoflurane.
Collapse
Affiliation(s)
- Hui Yuan
- Department of Anesthesiology, HwaMei Hospital, University of Chinese Academy of Sciences, Ningbo, China
| | - Guorong Wu
- Department of Anesthesiology, HwaMei Hospital, University of Chinese Academy of Sciences, Ningbo, China
| | - Xiaojie Zhai
- Department of Anesthesiology, HwaMei Hospital, University of Chinese Academy of Sciences, Ningbo, China
| | - Bo Lu
- Department of Anesthesiology, HwaMei Hospital, University of Chinese Academy of Sciences, Ningbo, China
| | - Bo Meng
- Department of Anesthesiology, HwaMei Hospital, University of Chinese Academy of Sciences, Ningbo, China
| | - Junping Chen
- Department of Anesthesiology, HwaMei Hospital, University of Chinese Academy of Sciences, Ningbo, China
| |
Collapse
|
25
|
Okamoto T, Kawashima H, Osada H, Toda E, Homma K, Nagai N, Imai Y, Tsubota K, Ozawa Y. Dietary Spirulina Supplementation Protects Visual Function From Photostress by Suppressing Retinal Neurodegeneration in Mice. Transl Vis Sci Technol 2019; 8:20. [PMID: 31788349 PMCID: PMC6871545 DOI: 10.1167/tvst.8.6.20] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2019] [Accepted: 08/29/2019] [Indexed: 01/01/2023] Open
Abstract
PURPOSE We investigated whether daily consumption of Spirulina, an antioxidant generating cyanobacterial nutritional supplement, would suppress photostress-induced retinal damage and prevent vision loss in mice. METHODS Six-week-old male BALB/cAJcl mice were allowed constant access to either a standard or Spirulina-supplemented diet (20% Spirulina) that included the antioxidants, β-carotene and zeaxanthin, and proteins for 4 weeks. Following dark adaptation, mice were exposed to 3000-lux white light for 1 hour and returned to their cages. Visual function was analyzed by electroretinogram, and retinal histology by hematoxylin and eosin staining, terminal deoxynucleotidyl transferase-mediated, deoxyuridine triphosphate nick-end labeling (TUNEL) assay, and immunohistochemistry. Retinal expression of proteins, reactive oxygen species (ROS), and mRNAs were measured using immunoblot analysis, enzyme-linked immunosorbent assay (ELISA), 2',7'-dichlorofluorescein-diacetate, or ROS Brite 700 Dyes, and real-time reverse-transcription polymerase chain reaction, respectively. RESULTS Light-induced visual function impairment was suppressed by constant Spirulina intake. Thinning of the photoreceptor layer and outer segments, photoreceptor cell death, decreased rhodopsin protein, and induction of glial fibrillary acidic protein were ameliorated in the Spirulina-intake group. Increased retinal ROS levels after light exposure were reduced by Spirulina supplementation. Light-induced superoxide dismutase 2 and heme oxygenase-1 mRNAs in the retina, and Nrf2 activation in the photoreceptor cells, were preserved with Spirulina supplementation, despite reduced ROS levels, suggesting two pathways for suppressing ROS, scavenging and induction of endogenous antioxidative enzymes. Light-induced MCP-1 retinal mRNA and proteins were also suppressed by Spirulina. CONCLUSIONS Spirulina ingestion protected retinal photoreceptors from photostress in the retina. TRANSLATIONAL RELEVANCE Spirulina has potential as a nutrient supplement to prevent vision loss related to oxidative damage in the future.
Collapse
Affiliation(s)
- Tomohiro Okamoto
- Laboratory of Retinal Cell Biology, Keio University School of Medicine, Tokyo, Japan
- Department of Ophthalmology, Keio University School of Medicine, Tokyo, Japan
| | - Hirohiko Kawashima
- Laboratory of Retinal Cell Biology, Keio University School of Medicine, Tokyo, Japan
- Department of Ophthalmology, Keio University School of Medicine, Tokyo, Japan
| | - Hideto Osada
- Laboratory of Retinal Cell Biology, Keio University School of Medicine, Tokyo, Japan
- Department of Ophthalmology, Keio University School of Medicine, Tokyo, Japan
| | - Eriko Toda
- Laboratory of Retinal Cell Biology, Keio University School of Medicine, Tokyo, Japan
- Department of Ophthalmology, Keio University School of Medicine, Tokyo, Japan
| | - Kohei Homma
- Laboratory of Retinal Cell Biology, Keio University School of Medicine, Tokyo, Japan
- Department of Ophthalmology, Keio University School of Medicine, Tokyo, Japan
| | - Norihiro Nagai
- Laboratory of Retinal Cell Biology, Keio University School of Medicine, Tokyo, Japan
- Department of Ophthalmology, Keio University School of Medicine, Tokyo, Japan
| | | | - Kazuo Tsubota
- Department of Ophthalmology, Keio University School of Medicine, Tokyo, Japan
| | - Yoko Ozawa
- Laboratory of Retinal Cell Biology, Keio University School of Medicine, Tokyo, Japan
- Department of Ophthalmology, Keio University School of Medicine, Tokyo, Japan
| |
Collapse
|
26
|
The uPAR System as a Potential Therapeutic Target in the Diseased Eye. Cells 2019; 8:cells8080925. [PMID: 31426601 PMCID: PMC6721659 DOI: 10.3390/cells8080925] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2019] [Revised: 08/07/2019] [Accepted: 08/17/2019] [Indexed: 12/15/2022] Open
Abstract
Dysregulation of vascular networks is characteristic of eye diseases associated with retinal cell degeneration and visual loss. Visual impairment is also the consequence of photoreceptor degeneration in inherited eye diseases with a major inflammatory component, but without angiogenic profile. Among the pathways with high impact on vascular/degenerative diseases of the eye, a central role is played by a system formed by the ligand urokinase-type plasminogen activator (uPA) and its receptor uPAR. The uPAR system, although extensively investigated in tumors, still remains a key issue in vascular diseases of the eye and even less studied in inherited retinal pathologies such as retinitis pigmantosa (RP). Its spectrum of action has been extended far beyond a classical pro-angiogenic function and has emerged as a central actor in inflammation. Preclinical studies in more prevalent eye diseases characterized by neovascular formation, as in retinopathy of prematurity, wet macular degeneration and rubeosis iridis or vasopermeability excess as in diabetic retinopathy, suggest a critical role of increased uPAR signaling indicating the potentiality of its modulation to counteract neovessel formation and microvascular dysfunction. The additional observation that the uPAR system plays a major role in RP by limiting the inflammatory cascade triggered by rod degeneration rises further questions about its role in the diseased eye.
Collapse
|
27
|
Wang JH, Wei ZF, Gao YL, Liu CC, Sun JH. Activation of the mammalian target of rapamycin signaling pathway underlies a novel inhibitory role of ring finger protein 182 in ventricular remodeling after myocardial ischemia-reperfusion injury. J Cell Biochem 2019; 120:7635-7648. [PMID: 30450663 DOI: 10.1002/jcb.28038] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2018] [Accepted: 10/15/2018] [Indexed: 01/24/2023]
Abstract
Myocardial ischemia-reperfusion injury (MIRI) is a major cause of cardiovascular disease, leading to mortality and disability associated with coronary occlusion worldwide. A correlation of mammalian target of rapamycin (mTOR)/nuclear factor-kappa B (NF-κB) signaling pathway has been observed with brain damage resulting from myocardial ischemia. Therefore, by establishing MIRI rat model, this study aimed to explore whether ring finger protein 182 (RNF182) regulates the mTOR signaling pathway affecting MIRI. Initially, MIRI rat model was successfully established, followed by either treatment of shRNF182 or phosphoesterase (PITE) (inhibitor of the mTOR signaling pathway). Then, the serum levels of superoxide dismutase (SOD), glutathione peroxidase (GSH-Px) and malondialdehyde (MDA), left ventricular ejection fraction (LVEF), left ventricular fractional shortening (LVFS), left ventricular systolic pressure (LVSP), and left ventricular end-diastolic pressure (LVEDP) were determined, followed by detection of myocardial infarct sizes and myocardial cell apoptosis. Moreover, the levels of related genes/proteins were determined to further determine the mechanisms of RNF182 in MIRI. First, RNF182 was upregulated in MIRI. Another key observation of this study was that rats with shRNF182 presented with downregulated SOD, GSH-Px, and MDA in serum, accompanied by decreased levels of LVEF, LVFS, LVSP, and LVEDP. In addition, both reduced myocardial infarct sizes and apoptosis of myocardial cells were observed after silencing RNF182. Furthermore, silencing of the RNF182 was observed to downregulate Bcl 2-associated X and cysteine proteinase 3 but upregulate mTOR, ribosome protein subunit 6 kinase 1, eukaryotic elongation factor 2, and B-cell lymphoma-2. Importantly, the effects of RNF182 silencing were reversed after PITE treatment. In conclusion, our study demonstrates that RNF182 silencing can prevent ventricular remodeling in rats after MIRI by activating the mTOR signaling pathway.
Collapse
Affiliation(s)
- Jing-Hua Wang
- Department of Pediatric Rheumatology, Immunology and Allergy, The First Hospital of Jilin University, Changchun, China
| | - Zhi-Feng Wei
- Department of Cardiology, FAW General Hospital, Changchun, China
| | - Yan-Li Gao
- Department of Science and Education, The First Hospital of Jilin University, Changchun, China
| | - Cong-Cong Liu
- Department of Pediatric Rheumatology, Immunology and Allergy, The First Hospital of Jilin University, Changchun, China
| | - Jing-Hui Sun
- Department of Pediatric Cardiology, The First Hospital of Jilin University, Changchun, China
| |
Collapse
|
28
|
Hu S, Cheng M, Fan R, Wang Z, Wang L, Zhang T, Zhang M, Louis E, Zhong J. Beneficial effects of dual TORC1/2 inhibition on chronic experimental colitis. Int Immunopharmacol 2019; 70:88-100. [PMID: 30797172 DOI: 10.1016/j.intimp.2019.02.022] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2018] [Revised: 02/12/2019] [Accepted: 02/12/2019] [Indexed: 12/14/2022]
Abstract
BACKGROUND AND AIM AZD8055, a new immunosuppressive reagent, a dual TORC1/2 inhibitor, had been used successfully in animal models for heart transplantation. The aim of this study was to evaluate the effects and mechanisms of AZD8055 on chronic intestinal inflammation. METHODS Dextran sulfate sodium (DSS) - induced chronic colitis was used to investigate the effects of AZD8055 on the development of colitis. Colitis activity was monitored by body weight assessment, colon length, histology and cytokine profile analysis. RESULTS AZD8055 treatment significantly alleviated the severity of colitis, as assessed by colonic length and colonic damage. In addition, AZD8055 treatment decreased the colonic CD4+ T cell numbers and reduced both Th1 and Th17 cell activation and cytokine production. The percentages of Treg cells in the colon were also expanded by AZD8055 treatment. Furthermore, AZD8055 effectively inhibited mTOR downstream proteins and signal transducer and activator of transcription related proteins in CD4+ T cells of intestinal lamina propria. CONCLUSIONS These findings increased our understanding of DSS-induced colitis and shed new lights on mechanisms of digestive tract chronic inflammation. Dual TORC1/2 inhibition showed potent anti-inflammatory and immune regulation effects by targeting critical signaling pathways. The results supported the strategy of using dual mTOR inhibitor to treat inflammatory bowel disease.
Collapse
Affiliation(s)
- Shurong Hu
- Department of Gastroenterology, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, 200025 Shanghai, PR China; Translational Gastroenterology Research Unit, GIGA-R, University of Liège, Belgium
| | - Mengmeng Cheng
- Department of Gastroenterology, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, 200025 Shanghai, PR China; Department of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and technology, Wuhan, Hubei, PR China
| | - Rong Fan
- Department of Gastroenterology, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, 200025 Shanghai, PR China
| | - Zhengting Wang
- Department of Gastroenterology, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, 200025 Shanghai, PR China
| | - Lei Wang
- Department of Gastroenterology, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, 200025 Shanghai, PR China
| | - Tianyu Zhang
- Department of Gastroenterology, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, 200025 Shanghai, PR China
| | - Maochen Zhang
- Department of Gastroenterology, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, 200025 Shanghai, PR China
| | - Edouard Louis
- Translational Gastroenterology Research Unit, GIGA-R, University of Liège, Belgium; Hepato-Gastroenterology and Digestive Oncology Unit, University Hospital, CHU Liege, Domaine du Sart Tilman, 4000 Liege, Belgium.
| | - Jie Zhong
- Department of Gastroenterology, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, 200025 Shanghai, PR China.
| |
Collapse
|
29
|
Shi B, Ma M, Zheng Y, Pan Y, Lin X. mTOR and Beclin1: Two key autophagy-related molecules and their roles in myocardial ischemia/reperfusion injury. J Cell Physiol 2019; 234:12562-12568. [PMID: 30618070 DOI: 10.1002/jcp.28125] [Citation(s) in RCA: 149] [Impact Index Per Article: 24.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2018] [Accepted: 12/18/2018] [Indexed: 12/12/2022]
Abstract
Autophagy is the general term of lysosomal degradation of substances in cells, which is considered the key to maintaining the normal structure and function of the heart. It also has a correlation with several heart diseases, in particular, myocardial ischemia/reperfusion (I/R) injury. At the stage of myocardial ischemia, autophagy degrades nonfunctional cytoplasmic proteins providing the critical nutrients for the critical life activities, thereby suppressing cell apoptosis and necrosis. However, autophagy is likely to affect the heart negatively in the reperfusion stage. Mammalian target of rapamycin (mTOR) and Beclin1 are two vital autophagy-related molecules in myocardial I/R injury playing significant roles in different stages. In the ischemia stage, mTOR plays its roles through AMPK/mTOR and phosphoinositide 3-kinase/Akt/mTOR pathway, whereas Beclin1 plays its roles through its upregulation in the reperfusion stage. A possible interaction between mTOR and Beclin1 has been reported recently, and further studies need to be done to find the underlying interaction between the two molecules in myocardial I/R injury.
Collapse
Affiliation(s)
- Binhao Shi
- Department of Cardiology, The First Affiliated Hospital of Anhui Medical University, Hefei, China.,Department of Clinical Medical, Anhui Medical University, Hefei, China
| | - Mengqing Ma
- Department of Cardiology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Yitian Zheng
- Department of Cardiology, The First Affiliated Hospital of Anhui Medical University, Hefei, China.,Department of Clinical Medical, Anhui Medical University, Hefei, China
| | - Yanyan Pan
- Department of Cardiology, The First Affiliated Hospital of Anhui Medical University, Hefei, China.,Department of Clinical Medical, Anhui Medical University, Hefei, China
| | - Xianhe Lin
- Department of Cardiology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| |
Collapse
|
30
|
Jia X, Cao B, An Y, Zhang X, Wang C. Rapamycin ameliorates lipopolysaccharide-induced acute lung injury by inhibiting IL-1β and IL-18 production. Int Immunopharmacol 2018; 67:211-219. [PMID: 30557824 DOI: 10.1016/j.intimp.2018.12.017] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2018] [Revised: 11/30/2018] [Accepted: 12/07/2018] [Indexed: 01/06/2023]
Abstract
Interleukin (IL)-1β and IL-18 play central and detrimental roles in the development of acute lung injury (ALI), and mammalian target of rapamycin (mTOR) is involved in regulating IL-1β and IL-18 production. However, it is not clear whether the mTOR specific inhibitor rapamycin can attenuate lipopolysaccharide (LPS)-induced ALI by modulating IL-1β and IL-18 production. In this study, we found that rapamycin ameliorated LPS-induced ALI by inhibiting NOD-like receptor family pyrin domain containing 3 (NLRP3) inflammasome-mediated IL-1β and IL-18 secretion. Mechanistically, elevated autophagy and decreased nuclear factor (NF)-κB activation were associated with downregulated IL-1β and IL-18. Moreover, rapamycin reduced leukocyte infiltration in the lung tissue and bronchoalveolar lavage fluid (BALF), and contributed to the alleviation of LPS-induced ALI. Consistently, rapamycin also significantly inhibited IL-1β and IL-18 production by RAW264.7 cells via increased autophagy and decreased NF-κB signaling in vitro. Our results demonstrated that rapamycin protects mice against LPS-induced ALI partly by inhibiting the production and secretion of IL-1β and IL-18. mTOR and rapamycin might represent an appropriate therapeutic target and strategy for preventing ALI induced by LPS.
Collapse
Affiliation(s)
- Xuehong Jia
- Department of Respiratory Medicine, Beijing Tiantan Hospital, Capital Medical University, Beijing 100050, China; Peking University China-Japan Friendship School of Clinical Medicine, Beijing 100029, China
| | - Bin Cao
- Department of Pulmonary and Critical Care Medicine, Center for Respiratory Diseases, China-Japan Friendship Hospital, Beijing 100029, China; National Clinical Research Center for Respiratory Diseases, Beijing 100029, China; Department of Respiratory Medicine, Capital Medical University, Beijing 100069, China
| | - Yunqing An
- Department of Immunology, School of Basic Medical Sciences, Capital Medical University, Beijing 100069, China
| | - Xulong Zhang
- Department of Immunology, School of Basic Medical Sciences, Capital Medical University, Beijing 100069, China.
| | - Chen Wang
- Peking University China-Japan Friendship School of Clinical Medicine, Beijing 100029, China; Department of Pulmonary and Critical Care Medicine, Center for Respiratory Diseases, China-Japan Friendship Hospital, Beijing 100029, China; National Clinical Research Center for Respiratory Diseases, Beijing 100029, China; Department of Respiratory Medicine, Capital Medical University, Beijing 100069, China; Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100730, China.
| |
Collapse
|
31
|
Delayed oseltamivir plus sirolimus treatment attenuates H1N1 virus-induced severe lung injury correlated with repressed NLRP3 inflammasome activation and inflammatory cell infiltration. PLoS Pathog 2018; 14:e1007428. [PMID: 30422993 PMCID: PMC6258564 DOI: 10.1371/journal.ppat.1007428] [Citation(s) in RCA: 68] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2018] [Revised: 11/27/2018] [Accepted: 10/22/2018] [Indexed: 12/23/2022] Open
Abstract
Severe influenza A virus infection causes high mortality and morbidity worldwide due to delayed antiviral treatment and inducing overwhelming immune responses, which contribute to immunopathological lung injury. Sirolimus, an inhibitor of mammalian target of rapamycin (mTOR), was effective in improving clinical outcomes in patients with severe H1N1 infection; however, the mechanisms by which it attenuates acute lung injury have not been elucidated. Here, delayed oseltamivir treatment was used to mimic clinical settings on lethal influenza A (H1N1) pdm09 virus (pH1N1) infection mice model. We revealed that delayed oseltamivir plus sirolimus treatment protects mice against lethal pH1N1 infection by attenuating severe lung damage. Mechanistically, the combined treatment reduced viral titer and pH1N1-induced mTOR activation. Subsequently, it suppressed the NOD-like receptor family pyrin domain containing 3 (NLRP3) inflammasome-mediated secretion of interleukin (IL)-1β and IL-18. It was noted that decreased NLRP3 inflammasome activation was associated with inhibited nuclear factor (NF)-κB activation, reduced reactive oxygen species production and increased autophagy. Additionally, the combined treatment reduced the expression of other proinflammatory cytokines and chemokines, and decreased inflammatory cell infiltration in lung tissue and bronchioalveolar lavage fluid. Consistently, it inhibited the mTOR-NF-κB-NLRP3 inflammasome-IL-1β axis in a lung epithelial cell line. These results demonstrated that combined treatment with sirolimus and oseltamivir attenuates pH1N1-induced severe lung injury, which is correlated with suppressed mTOR-NLRP3-IL-1β axis and reduced viral titer. Therefore, treatment with sirolimus as an adjuvant along with oseltamivir may be a promising immunomodulatory strategy for managing severe influenza. The severity and lethality of influenza A virus infection are frequently aggravated by virus-induced tissue destruction and overwhelming immune responses. Combined therapy with antiviral medications and immunomodulators, which not only inhibit viral replication, but also reduce the damaging consequences of host immune responses, will be beneficial in the treatment of severe influenza. In the present study, we revealed that pH1N1-induced activation of mTOR promotes lung immunopathological injury, which is correlated with upregulated NF-κB activity and increased reactive oxygen species production. Subsequently, it induces NLRP3 inflammasome activation and the secretion of IL-1β and IL-18. Combined treatment with oseltamivir and the mTOR inhibitor sirolimus (as an adjuvant) not only blocks viral replication, but also suppresses mTOR-NLRP3-IL-1β axis-mediated immune damage, thus protecting mice against lethal pH1N1 infection. Our findings provide the theoretical and experimental basis for the clinical investigation of sirolimus as an adjunct treatment for severe influenza.
Collapse
|
32
|
Xu R, Lin J, Zhao GQ, Li C, Che CY, Xu Q, Liu M. Production of interleukin-1β related to mammalian target of rapamycin/Toll-like receptor 4 signaling pathway during Aspergillus fumigatus infection of the mouse cornea. Int J Ophthalmol 2018; 11:712-718. [PMID: 29862167 DOI: 10.18240/ijo.2018.05.02] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2017] [Accepted: 03/19/2018] [Indexed: 12/15/2022] Open
Abstract
AIM To elucidate the effect of rapamycin on regulating the production of interleukin (IL)-1β in Aspergillus fumigatus (A. fumigatus)-induced keratitis and to verify whether the expression of IL-1β in A. fumigatus keratitis is associated with the mammalian target of rapamycin (mTOR)/Toll-like receptor 4 (TLR4) signaling pathway. METHODS Fungal keratitis mouse models of susceptible C57BL/6 mice were established using A. fumigatus. The mice were subsequently treated with rapamycin. The protein levels of p-mTOR, TLR4, and IL-1β in normal and infected corneal tissue were measured by Western blot. The TLR4 and IL-1β mRNA levels were determined by real-time polymerase chain reaction (PCR). RESULTS In C57BL/6 mice, rapamycin treatment decreased the clinical scores and production of the pro-inflammatory cytokine, IL-1β. The expression of TLR4, stimulated by A. fumigatus, was reduced as well when the mTOR signaling pathway was suppressed by rapamycin. CONCLUSION Rapamycin is beneficial for the outcome of fungal keratitis and has an inhibitory effect expression of the inflammatory cytokine IL-1β. The inhibitory effect on IL-1β expression can be associated with the mTOR/TLR4 signaling pathway in A. fumigatus infection in mice.
Collapse
Affiliation(s)
- Rui Xu
- Department of Ophthalmology, the Affiliated Hospital of Qingdao University, Qingdao 266003, Shandong Province, China
| | - Jing Lin
- Department of Ophthalmology, the Affiliated Hospital of Qingdao University, Qingdao 266003, Shandong Province, China
| | - Gui-Qiu Zhao
- Department of Ophthalmology, the Affiliated Hospital of Qingdao University, Qingdao 266003, Shandong Province, China
| | - Cui Li
- Department of Ophthalmology, the Affiliated Hospital of Qingdao University, Qingdao 266003, Shandong Province, China
| | - Cheng-Ye Che
- Department of Ophthalmology, the Affiliated Hospital of Qingdao University, Qingdao 266003, Shandong Province, China
| | - Qiang Xu
- Department of Ophthalmology, the Affiliated Hospital of Qingdao University, Qingdao 266003, Shandong Province, China
| | - Min Liu
- Department of Ophthalmology, the Affiliated Hospital of Qingdao University, Qingdao 266003, Shandong Province, China
| |
Collapse
|
33
|
Huang W, Wen J, Lin R, Wei P, Huang F. Effects of mTOR/NF‐κB signaling pathway and high thoracic epidural anesthesia on myocardial ischemia‐reperfusion injury via autophagy in rats. J Cell Physiol 2018; 233:6669-6678. [DOI: 10.1002/jcp.26320] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2017] [Accepted: 11/30/2017] [Indexed: 01/21/2023]
Affiliation(s)
- Wei‐Qiang Huang
- Cardio‐Cerebrovascular Disease Precision Medical Key Laboratory Cultivation Base of GuangxiDepartment of Geriatric CardiologyThe First Affiliated Hospital of Guangxi Medical UniversityNanningChina
| | - Jian‐Lin Wen
- Cardio‐Cerebrovascular Disease Precision Medical Key Laboratory Cultivation Base of GuangxiDepartment of Geriatric CardiologyThe First Affiliated Hospital of Guangxi Medical UniversityNanningChina
| | - Ri‐Qi Lin
- Cardio‐Cerebrovascular Disease Precision Medical Key Laboratory Cultivation Base of GuangxiDepartment of Geriatric CardiologyThe First Affiliated Hospital of Guangxi Medical UniversityNanningChina
| | - Peng Wei
- Cardio‐Cerebrovascular Disease Precision Medical Key Laboratory Cultivation Base of GuangxiDepartment of Geriatric CardiologyThe First Affiliated Hospital of Guangxi Medical UniversityNanningChina
| | - Feng Huang
- Cardio‐Cerebrovascular Disease Precision Medical Key Laboratory Cultivation Base of GuangxiDepartment of CardiologyThe First Affiliated Hospital of Guangxi Medical UniversityNanningChina
| |
Collapse
|
34
|
Xu Y, Yang B, Hu Y, Lu L, Lu X, Wang J, Xu F, Yu S, Huang J, Liang X. Wogonin prevents TLR4-NF-κB-medicated neuro-inflammation and improves retinal ganglion cells survival in retina after optic nerve crush. Oncotarget 2018; 7:72503-72517. [PMID: 27756890 PMCID: PMC5341925 DOI: 10.18632/oncotarget.12700] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2016] [Accepted: 10/05/2016] [Indexed: 01/11/2023] Open
Abstract
Chronic neuro-inflammation is involved in the death of retinal ganglion cells (RGCs) in glaucoma. The aim of this study is to determine whether wogonin can suppress inflammatory responses and rescue RGCs death after optic nerve crush (ONC), an ideal animal model of glaucoma. Wogonin was administered intraperitoneally 10 min after establishment of ONC model. In this study, wogonin treatment reduced RGCs loss and inhibited RGCs apoptosis demonstrated by the increased Brn3a labeling RGCs at day 14 and the decreased cleaved caspase-3 expression at day 7 after ONC, respectively. In ONC model, number of GFAP-positive glial cells and iba1-positive microglial cells were increased, combined of the elevated level of pro-inflammatory cytokines released in retina at day 7. However, most of these responses were inhibited after wogonin treatment. The level of TLR4 expression, NF-κB-P65 nucleus location and NF-κB-P65 phosphorylation were increased in retina at day 1 after ONC, which was significantly reduced after wogonin treatment. These results demonstrated that wogonin protected RGCs survival and suppressed neuro-inflammation in retina after ONC by inhibiting TLR4-NF-κB pathways. We conclude that wogonin could be a possible strategy for the treatment of glaucoma.
Collapse
Affiliation(s)
- Yue Xu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, Guangdong, People's Republic of China
| | - Boyu Yang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, Guangdong, People's Republic of China
| | - Yaguang Hu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, Guangdong, People's Republic of China
| | - Lin Lu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, Guangdong, People's Republic of China
| | - Xi Lu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, Guangdong, People's Republic of China
| | - Jiawei Wang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, Guangdong, People's Republic of China
| | - Fan Xu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, Guangdong, People's Republic of China
| | - Shanshan Yu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, Guangdong, People's Republic of China
| | - Jingjing Huang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, Guangdong, People's Republic of China
| | - Xiaoling Liang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, Guangdong, People's Republic of China
| |
Collapse
|
35
|
Donato L, Bramanti P, Scimone C, Rinaldi C, Giorgianni F, Beranova-Giorgianni S, Koirala D, D'Angelo R, Sidoti A. miRNAexpression profile of retinal pigment epithelial cells under oxidative stress conditions. FEBS Open Bio 2018; 8:219-233. [PMID: 29435412 PMCID: PMC5794457 DOI: 10.1002/2211-5463.12360] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2017] [Revised: 11/03/2017] [Accepted: 11/24/2017] [Indexed: 12/22/2022] Open
Abstract
Deep analysis of regulative mechanisms of transcription and translation in eukaryotes could improve knowledge of many genetic pathologies such as retinitis pigmentosa (RP). New layers of complexity have recently emerged with the discovery that ‘junk’ DNA is transcribed and, among these, miRNAs have assumed a preponderant role. We compared changes in the expression of miRNAs obtained from whole transcriptome analyses, between two groups of retinal pigment epithelium (RPE) cells, one untreated and the other exposed to the oxidant agent oxidized low‐density lipoprotein (oxLDL), examining four time points (1, 2, 4 and 6 h). We found that 23 miRNAs exhibited altered expression in the treated samples, targeting genes involved in several biochemical pathways, many of them associated to RP for the first time, such as those mediated by insulin receptor signaling and son of sevenless. Moreover, five RP causative genes (KLHL7, RDH11,CERKL, AIPL1 and USH1G) emerged as already validated targets of five altered miRNAs (hsa‐miR‐1307, hsa‐miR‐3064, hsa‐miR‐4709, hsa‐miR‐3615 and hsa‐miR‐637), suggesting a tight connection between induced oxidative stress and RP development and progression. This miRNA expression analysis of oxidative stress‐induced RPE cells has discovered new regulative functions of miRNAs in RP that should lead to the discovery of new ways to regulate the etiopathogenesis of RP.
Collapse
Affiliation(s)
- Luigi Donato
- Department of Biomedical and Dental Sciences and Morphofunctional Imaging Division of Medical Biotechnologies and Preventive Medicine University of Messina Italy.,Department of Cutting-Edge Medicine and Therapies Biomolecular Strategies and Neuroscience Section of Neuroscience-applied, Molecular Genetics and Predictive MedicineI.E.M E.S.T. Palermo Italy
| | | | - Concetta Scimone
- Department of Biomedical and Dental Sciences and Morphofunctional Imaging Division of Medical Biotechnologies and Preventive Medicine University of Messina Italy.,Department of Cutting-Edge Medicine and Therapies Biomolecular Strategies and Neuroscience Section of Neuroscience-applied, Molecular Genetics and Predictive MedicineI.E.M E.S.T. Palermo Italy
| | - Carmela Rinaldi
- Department of Biomedical and Dental Sciences and Morphofunctional Imaging Division of Medical Biotechnologies and Preventive Medicine University of Messina Italy
| | | | | | | | - Rosalia D'Angelo
- Department of Biomedical and Dental Sciences and Morphofunctional Imaging Division of Medical Biotechnologies and Preventive Medicine University of Messina Italy
| | - Antonina Sidoti
- Department of Biomedical and Dental Sciences and Morphofunctional Imaging Division of Medical Biotechnologies and Preventive Medicine University of Messina Italy.,Department of Cutting-Edge Medicine and Therapies Biomolecular Strategies and Neuroscience Section of Neuroscience-applied, Molecular Genetics and Predictive MedicineI.E.M E.S.T. Palermo Italy
| |
Collapse
|
36
|
Kim TW, Han JM, Han YK, Chung H. Anti-inflammatory Effects of Sinomenium Acutum Extract On Endotoxin-induced Uveitis in Lewis Rats. Int J Med Sci 2018; 15:758-764. [PMID: 30008584 PMCID: PMC6036085 DOI: 10.7150/ijms.24834] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2018] [Accepted: 04/27/2018] [Indexed: 01/08/2023] Open
Abstract
PURPOSE Recent studies have reported the anti-inflammatory effect of Sinomenium acutum. We investigated the anti-inflammatory effect of sinomenine on endotoxin-induced uveitis in a rat model. METHODS Endotoxin-induced uveitis was induced in rat by lipopolysaccharide (LPS) immunization. Sinomenine (50mg/kg and 100mg/kg) was administered at 30 minutes before, 6 hours and 12 hours after LPS immunization. Clinical and histological severity was evaluated. Protein concentration and levels of tumor necrosis factor (TNF)-α and prostaglandin (PG)-E2 in aqueous humor were measured. Expression of activated Nuclear factor (NF)-κB p65 in ciliary body was also observed. RESULTS Clinical and histological severities were significantly milder in sinomenine-treated rat than in controls (P < 0.001). Sinomenine suppressed protein leakage and down-regulated the production of TNF-α and PG-E2 in a dose-dependent manner. Sinomenine treatment suppressed the translocation of the NF-κB p65 subunit into the nuclei. CONCLUSION Systemic administration of sinomenine suppressed the inflammation of ocular tissues. These findings suggest that sinomenine could be a novel therapeutic agent for the control of endogenous ocular inflammatory disease.
Collapse
Affiliation(s)
- Tae Wan Kim
- Department of Ophthalmology, Seoul Metropolitan Government Seoul National University Boramae Medical Center, Seoul, Korea.,Department of Ophthalmology, Seoul National University College of Medicine, Seoul, Korea
| | - Jeong Mo Han
- Department of Ophthalmology, Seoul National University College of Medicine, Seoul, Korea
| | - Young Keun Han
- Department of Ophthalmology, Seoul Metropolitan Government Seoul National University Boramae Medical Center, Seoul, Korea
| | - Hokyung Chung
- Department of Ophthalmology, Seoul Metropolitan Government Seoul National University Boramae Medical Center, Seoul, Korea
| |
Collapse
|
37
|
Soltani A, Bahreyni A, Boroumand N, Roshan MK, Khazaei M, Ryzhikov M, Soleimanpour S, Avan A, Hassanian SM. Therapeutic potency of mTOR signaling pharmacological inhibitors in the treatment of proinflammatory diseases, current status, and perspectives. J Cell Physiol 2017; 233:4783-4790. [PMID: 29165795 DOI: 10.1002/jcp.26276] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2017] [Revised: 10/14/2017] [Accepted: 11/14/2017] [Indexed: 12/26/2022]
Abstract
Mammalian target of rapamycin (mTOR) signaling pathway controls cell energy metabolism. There is an interplay between mTOR and proinflammatory signaling pathways, supporting the role of the pathway in the pathogenesis of inflammatory diseases. Inhibition of mTOR signaling using specific pharmacological inhibitors could offer therapeutic promise in several inflammatory-associated diseases. In this review, we summarize recent findings on the regulatory effects of mTOR signaling on inflammation and the therapeutic potency of mTOR pharmacological inhibitors in the treatment of inflammatory diseases including cancer, neurodegenerative diseases, atherosclerosis, sepsis, and rheumatoid arthritis for a better understanding and hence a better management of these diseases.
Collapse
Affiliation(s)
- Arash Soltani
- Faculty of Medicine, Department of Medical Biochemistry, Mashhad University of Medical Sciences, Mashhad, Iran.,Student Research Committee, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Amirhossein Bahreyni
- Department of Clinical Biochemistry and Immunogenetic Research Center, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Mazandaran, Iran
| | - Nadia Boroumand
- Faculty of Medicine, Department of Medical Biochemistry, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mostafa Karimi Roshan
- Faculty of Medicine, Department of Medical Biochemistry, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Majid Khazaei
- Faculty of Medicine, Department of Medical Physiology, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mikhail Ryzhikov
- Department of Molecular Microbiology and Immunology, St. Louis University, School of Medicine, Saint Louis, Missouri
| | - Saman Soleimanpour
- Department of Microbiology and Virology, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Amir Avan
- Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.,Faculty of Medicine, Department of Modern Sciences and Technologies, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Seyed Mahdi Hassanian
- Faculty of Medicine, Department of Medical Biochemistry, Mashhad University of Medical Sciences, Mashhad, Iran.,Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.,Faculty of Medicine, Department of Modern Sciences and Technologies, Mashhad University of Medical Sciences, Mashhad, Iran.,Microanatomy Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
38
|
Cammalleri M, Locri F, Catalani E, Filippi L, Cervia D, Dal Monte M, Bagnoli P. The Beta Adrenergic Receptor Blocker Propranolol Counteracts Retinal Dysfunction in a Mouse Model of Oxygen Induced Retinopathy: Restoring the Balance between Apoptosis and Autophagy. Front Cell Neurosci 2017; 11:395. [PMID: 29375312 PMCID: PMC5770647 DOI: 10.3389/fncel.2017.00395] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2017] [Accepted: 11/28/2017] [Indexed: 12/29/2022] Open
Abstract
In a mouse model of oxygen induced retinopathy (OIR), beta adrenergic receptor (BAR) blockade has been shown to recover hypoxia-associated retinal damages. Although the adrenergic signaling is an important regulator of apoptotic and autophagic processes, the role of BARs in retinal cell death remains to be elucidated. The present study was aimed at investigating whether ameliorative effects of BAR blockers may occur through their coordinated action on apoptosis and autophagy. To this aim, retinas from control and OIR mice untreated or treated with propranolol, a non-selective BAR1/2 blocker, were characterized in terms of expression and localization of apoptosis and autophagy markers. The effects of propranolol on autophagy signaling were also evaluated and specific autophagy modulators were used to get functional information on the autophagic effects of BAR antagonism. Finally, propranolol effects on neurodegenerative processes were associated to an electrophysiological investigation of retinal function by recording electroretinogram (ERG). We found that retinas of OIR mice are characterized by increased apoptosis and decreased autophagy, while propranolol reduces apoptosis and stimulates autophagy. In particular, propranolol triggers autophagosome formation in bipolar, amacrine and ganglion cells that are committed to die by apoptosis in response to hypoxia. Also our data argue that propranolol, through the inhibition of the Akt-mammalian target of rapamycin pathway, activates autophagy which decreases retinal cell death. At the functional level, propranolol recovers dysfunctional ERG by recovering the amplitude of a- and b-waves, and oscillatory potentials, thus indicating an efficient restoring of retinal transduction. Overall, our results demonstrate that BAR1/2 are key regulators of retinal apoptosis/autophagy, and that BAR1/2 blockade leads to autophagy-mediated neuroprotection. Reinstating the balance between apoptotic and autophagic machines may therefore be viewed as a future goal in the treatment of retinopathies.
Collapse
Affiliation(s)
| | - Filippo Locri
- Department of Biology, University of Pisa, Pisa, Italy
| | - Elisabetta Catalani
- Department for Innovation in Biological, Agro-Food and Forest Systems, University of Tuscia, Viterbo, Italy
| | - Luca Filippi
- Neonatal Intensive Care Unit, Medical Surgical Fetal-Neonatal Department, Meyer University Children's Hospital, Florence, Italy
| | - Davide Cervia
- Department for Innovation in Biological, Agro-Food and Forest Systems, University of Tuscia, Viterbo, Italy
| | | | - Paola Bagnoli
- Department of Biology, University of Pisa, Pisa, Italy
| |
Collapse
|
39
|
Vanlandingham PA, Nuno DJ, Quiambao AB, Phelps E, Wassel RA, Ma JX, Farjo KM, Farjo RA. Inhibition of Stat3 by a Small Molecule Inhibitor Slows Vision Loss in a Rat Model of Diabetic Retinopathy. Invest Ophthalmol Vis Sci 2017; 58:2095-2105. [PMID: 28395025 PMCID: PMC5386345 DOI: 10.1167/iovs.16-20641] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Purpose Diabetic retinopathy is a leading cause of vision loss. Previous studies have shown signaling pathways mediated by Stat3 (signal transducer and activator of transcription 3) play a primary role in diabetic retinopathy progression. This study tested CLT-005, a small molecule inhibitor of Stat3, for its dose-dependent therapeutic effects on vision loss in a rat model of diabetic retinopathy. Methods Brown Norway rats were administered streptozotocin (STZ) to induce diabetes. CLT-005 was administered daily by oral gavage for 16 weeks at concentrations of 125, 250, or 500 mg/kg, respectively, beginning 4 days post streptozotocin administration. Systemic and ocular drug concentration was quantified with mass spectrometry. Visual function was monitored at 2-week intervals from 6 to 16 weeks using optokinetic tracking to measure visual acuity and contrast sensitivity. The presence and severity of cataracts was visually monitored and correlated to visual acuity. The transcription and translation of multiple angiogenic factors and inflammatory cytokines were measured by real-time polymerase chain reaction and Multiplex immunoassay. Results Streptozotocin-diabetic rats sustain progressive vision loss over 16 weeks, and this loss in visual function is rescued in a dose-dependent manner by CLT-005. This positive therapeutic effect correlates to the positive effects of CLT-005 on vascular leakage and the presence of inflammatory cytokines in the retina. Conclusions The present study indicates that Stat3 inhibition has strong therapeutic potential for the treatment of vision loss in diabetic retinopathy.
Collapse
Affiliation(s)
| | - Didier J Nuno
- Charlesson LLC, Oklahoma City, Oklahoma, United States
| | | | - Eric Phelps
- Charlesson LLC, Oklahoma City, Oklahoma, United States
| | - Ronald A Wassel
- Charlesson LLC, Oklahoma City, Oklahoma, United States 2EyeCRO, LLC, Oklahoma City, Oklahoma, United States
| | - Jian-Xing Ma
- Department of Physiology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, United States
| | - Krysten M Farjo
- Department of Physiology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, United States
| | - Rafal A Farjo
- Charlesson LLC, Oklahoma City, Oklahoma, United States 2EyeCRO, LLC, Oklahoma City, Oklahoma, United States
| |
Collapse
|
40
|
Osada H, Okamoto T, Kawashima H, Toda E, Miyake S, Nagai N, Kobayashi S, Tsubota K, Ozawa Y. Neuroprotective effect of bilberry extract in a murine model of photo-stressed retina. PLoS One 2017; 12:e0178627. [PMID: 28570634 PMCID: PMC5453571 DOI: 10.1371/journal.pone.0178627] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2017] [Accepted: 05/16/2017] [Indexed: 12/03/2022] Open
Abstract
Excessive exposure to light promotes degenerative and blinding retinal diseases such as age-related macular degeneration and retinitis pigmentosa. However, the underlying mechanisms of photo-induced retinal degeneration are not fully understood, and a generalizable preventive intervention has not been proposed. Bilberry extract is an antioxidant-rich supplement that ameliorates ocular symptoms. However, its effects on photo-stressed retinas have not been clarified. In this study, we examined the neuroprotective effects of bilberry extract against photo-stress in murine retinas. Light-induced visual function impairment recorded by scotopic and phototopic electroretinograms showing respective rod and cone photoreceptor function was attenuated by oral administration of bilberry extract through a stomach tube in Balb/c mice (750 mg/kg body weight). Bilberry extract also suppressed photo-induced apoptosis in the photoreceptor cell layer and shortening of the outer segments of rod and cone photoreceptors. Levels of photo-induced reactive oxygen species (ROS), oxidative and endoplasmic reticulum (ER) stress markers, as measured by real-time reverse transcriptase polymerase chain reaction, were reduced by bilberry extract treatment. Reduction of ROS by N-acetyl-L-cysteine, a well-known antioxidant also suppressed ER stress. Immunohistochemical analysis of activating transcription factor 4 expression showed the presence of ER stress in the retina, and at least in part, in Müller glial cells. The photo-induced disruption of tight junctions in the retinal pigment epithelium was also attenuated by bilberry extract, repressing an oxidative stress marker, although ER stress markers were not repressed. Our results suggest that bilberry extract attenuates photo-induced apoptosis and visual dysfunction most likely, and at least in part, through ROS reduction, and subsequent ER stress attenuation in the retina. This study can help understand the mechanisms of photo-stress and contribute to developing a new, potentially useful therapeutic approach using bilberry extract for preventing retinal photo-damage.
Collapse
Affiliation(s)
- Hideto Osada
- Laboratory of Retinal Cell Biology, Department of Ophthalmology, Keio University School of Medicine, Tokyo, Japan
| | - Tomohiro Okamoto
- Laboratory of Retinal Cell Biology, Department of Ophthalmology, Keio University School of Medicine, Tokyo, Japan
- Department of Ophthalmology, Keio University School of Medicine, Tokyo, Japan
| | - Hirohiko Kawashima
- Laboratory of Retinal Cell Biology, Department of Ophthalmology, Keio University School of Medicine, Tokyo, Japan
- Department of Ophthalmology, Keio University School of Medicine, Tokyo, Japan
| | - Eriko Toda
- Laboratory of Retinal Cell Biology, Department of Ophthalmology, Keio University School of Medicine, Tokyo, Japan
| | - Seiji Miyake
- Laboratory of Retinal Cell Biology, Department of Ophthalmology, Keio University School of Medicine, Tokyo, Japan
- Wakasa Seikatsu Co., Ltd., Kyoto, Japan
| | - Norihiro Nagai
- Laboratory of Retinal Cell Biology, Department of Ophthalmology, Keio University School of Medicine, Tokyo, Japan
- Department of Ophthalmology, Keio University School of Medicine, Tokyo, Japan
| | | | - Kazuo Tsubota
- Department of Ophthalmology, Keio University School of Medicine, Tokyo, Japan
| | - Yoko Ozawa
- Laboratory of Retinal Cell Biology, Department of Ophthalmology, Keio University School of Medicine, Tokyo, Japan
- Department of Ophthalmology, Keio University School of Medicine, Tokyo, Japan
- * E-mail:
| |
Collapse
|
41
|
Liu YC, Gao XX, Chen L, You XQ. Rapamycin suppresses Aβ 25-35- or LPS-induced neuronal inflammation via modulation of NF-κB signaling. Neuroscience 2017; 355:188-199. [PMID: 28504198 DOI: 10.1016/j.neuroscience.2017.05.005] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2016] [Revised: 04/25/2017] [Accepted: 05/03/2017] [Indexed: 01/10/2023]
Abstract
Rapamycin (RAPA), an inhibitor of mammalian target of rapamycin (mTOR), exhibits a high neuroprotective action against neurodegenerative diseases in mouse models. Since neuroinflammation has been shown to be involved in Alzheimer's disease (AD) development and progression, the aim of this study was to examine the anti-inflammatory role of RAPA in AD in vivo and in vitro, and investigate the underlying mechanisms. We found that amyloid-β (Aβ) induced neuronal inflammation and a remarkable increase in mTOR activity in in-vivo and in-vitro models of inflammation, suggesting the critical role of mTOR signaling in neuronal inflammation. In addition, administration of RAPA was found to down-regulate mTOR, p-mTOR, Nuclear factor kappa B (NF-κB) p65, p-p65, TNF-α, IL-1β and Bax protein expression in Aβ25-35- or lipopolysaccharides (LPS)-treated mice and cultured Neuro-2a (N2a) cells. Moreover, RAPA disrupted Aβ25-35-induced nuclear translocation of mTOR and NF-κB. Our findings indicate that RAPA inhibits Aβ25-35- or LPS-induced neuronal inflammation through suppressing mTOR signaling and reducing nuclear import of NF-κB.
Collapse
Affiliation(s)
- Ying-Chun Liu
- Department of Cell Biology and Genetics, School of Basic Medical Sciences, Fujian Medical University, No. 1 Xueyuan Road, Shangjie Town, Minhou County, Fuzhou City, Fujian Province 350108, China
| | - Xiao-Xiao Gao
- Department of Cell Biology and Genetics, School of Basic Medical Sciences, Fujian Medical University, No. 1 Xueyuan Road, Shangjie Town, Minhou County, Fuzhou City, Fujian Province 350108, China
| | - Ling Chen
- Department of Cell Biology and Genetics, School of Basic Medical Sciences, Fujian Medical University, No. 1 Xueyuan Road, Shangjie Town, Minhou County, Fuzhou City, Fujian Province 350108, China
| | - Xiao-Qing You
- Department of Cell Biology and Genetics, School of Basic Medical Sciences, Fujian Medical University, No. 1 Xueyuan Road, Shangjie Town, Minhou County, Fuzhou City, Fujian Province 350108, China.
| |
Collapse
|
42
|
Sirt1 ameliorates systemic sclerosis by targeting the mTOR pathway. J Dermatol Sci 2017; 87:149-158. [PMID: 28532580 DOI: 10.1016/j.jdermsci.2017.04.013] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2016] [Revised: 03/26/2017] [Accepted: 04/25/2017] [Indexed: 01/17/2023]
Abstract
BACKGROUND Systemic sclerosis (SSc) is a chronic autoimmune disease characterized by inflammation and fibrosis. Our previous research has indicated that Sirtuin1 (Sirt1) plays a role in the regulation of TNF-α-induced inflammation; however, whether Sirt1 may inhibit the progress of SSc by blocking inflammation remains unknown. OBJECTIVE We aimed to investigate the function of Sirt1 in SSc. METHODS The function and its mechanism of Sirt1 were evaluated in fibroblasts or scleroderma mice. The expression of Sirt1 and cytokines was analyzed using real-time PCR, western blot, ELISA and immunohistochemistry. RESULTS We determined that fibroblasts of SSc patients were activated to exhibit inflammation. Sirt1, activated by resveratrol (Res), ameliorated cutaneous inflammation and fibrosis in bleomycin (BLM)-induced scleroderma mice. An improvement in mammalian target of rapamycin (mTOR) was identified in the fibroblasts of SSc patients and the skin lesions of BLM mice. Rapamycin, an mTOR specific inhibitor, substantially inhibited the induced inflammation and fibrosis. The enhancement of mTOR expression in the skin lesions of the BLM-treated mice was significantly inhibited by Sirt1 activation. However, in both the BLM-treated cells and mice, Res exerted an inhibitory function on the expression of inflammatory factors, and collagen was diminished following mTOR knockdown. These findings suggest that Res may inhibit inflammation and fibrosis via mTOR. CONCLUSION The modulation of Sirt1 activity may represent a potential therapeutic method for SSc. The mechanism may involve the inhibition of mTOR phosphorylation, whereas mTOR activity was shown to be a pathogenic culprit of SSc.
Collapse
|
43
|
Yan WM, Zhang L, Chen T, Zhao GH, Long P, An J, Zhang ZM. Effects of hydrogen-rich saline on endotoxin-induced uveitis. Med Gas Res 2017; 7:9-18. [PMID: 28480027 PMCID: PMC5402351 DOI: 10.4103/2045-9912.202905] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
The therapeutic effects of hydrogen-rich saline (HRS) have been reported for a wide range of diseases mainly via selectively reducing the amount of reactive oxygen species. Oxidative stress plays an important role in the pathogenesis of uveitis and endotoxin-induced uveitis (EIU). In this study, we investigated whether HRS can mitigate EIU in rats. Sprague-Dawley rats were randomly divided into Norm group, Model group, HRS group, dexamethasone (DEX) group, and rats in the latter three groups were injected with equal amount of lipopolysaccharide (LPS) to induce EIU of different severities (by 1 mg/kg of LPS, or 1/8 mg/kg of LPS). Rats in HRS group were injected with HRS intraperitoneally at three different modes to purse an ameliorating effect of EIU (10 mL/kg of HRS immediately after injection of 1 mg/kg of LPS, 20 mL/kg of HRS once a day for 1 week before injection of 1 mg/kg of LPS and at 0, 0.5, 1, 2, 6, 8, 12 hours after LPS administration, or 20 mL/kg of HRS once a day for 1 week before injection of 1/8 mg/kg of LPS, and at 0, 0.5, 1, 2, 6, 8, 12, 24 hours and once a day for 3 weeks after LPS administration). Rats of DEX group were injected with 1 mL/kg of DEX solution intraperitoneally immediately after LPS administration. Rats in Norm and Model groups did not receive any treatment. All rats were examined under slit lamp microscope and graded according to the clinical signs of uveitis. Electroretinogram, quantitative analysis of protein in aqueous humor (AqH) and histological examination of iris and ciliary body were also carried out. Our results showed that HRS did not obviously ameliorate the signs of uveitis under slit lamp examination and the inflammatory cells infiltration around iris and cilliary body of EIU induced by 1 mg/kg or 1/8 mg/kg of LPS (P > 0.05), while DEX significantly reduced the inflammation reflected by the above two indicators (P < 0.05). The impaired retinal function of mild EIU induced by 1/8 mg/kg of LPS, showed by delay of peak time of b-wave of Dark adapted 3.0 electroretinogram, was not significantly restored by HRS (P > 0.05), while DEX had an obvious therapeutic effect (P < 0.05). However, HRS exerted an inhibition trend on elevation of protein in AqH of EIU induced by 1 mg/kg of LPS, and significantly reduced the increasing amount of protein in AqH of mild EIU induced by 1/8 mg/kg of LPS (P < 0.05). In conclusion, HRS could not obviously mitigate EIU in rats, while it could inhibit the elevation of AqH protein.
Collapse
Affiliation(s)
- Wei-ming Yan
- Department of Clinical Medicine, Faculty of Aerospace Medicine, Key Laboratory of Aerospace Medicine of the National Education Ministry, Fourth Military University, Xi’an, Shaanxi Province, China
| | - Lei Zhang
- Department of Clinical Medicine, Faculty of Aerospace Medicine, Key Laboratory of Aerospace Medicine of the National Education Ministry, Fourth Military University, Xi’an, Shaanxi Province, China
| | - Tao Chen
- Department of Health Service, Faculty of Aerospace Medicine, Key Laboratory of Aerospace Medicine of the National Education Ministry, Fourth Military University, Xi’an, Shaanxi Province, China
| | - Guan-hua Zhao
- Department of Clinical Medicine, Faculty of Aerospace Medicine, Key Laboratory of Aerospace Medicine of the National Education Ministry, Fourth Military University, Xi’an, Shaanxi Province, China
| | - Pan Long
- Department of Clinical Medicine, Faculty of Aerospace Medicine, Key Laboratory of Aerospace Medicine of the National Education Ministry, Fourth Military University, Xi’an, Shaanxi Province, China
| | - Jing An
- Institute of Neurobiology, School of Basic Medical Sciences, Xi’an Jiaotong University, Xi’an, Shaanxi Province, China
| | - Zuo-ming Zhang
- Department of Clinical Medicine, Faculty of Aerospace Medicine, Key Laboratory of Aerospace Medicine of the National Education Ministry, Fourth Military University, Xi’an, Shaanxi Province, China
| |
Collapse
|
44
|
Boya P, Esteban-Martínez L, Serrano-Puebla A, Gómez-Sintes R, Villarejo-Zori B. Autophagy in the eye: Development, degeneration, and aging. Prog Retin Eye Res 2016; 55:206-245. [PMID: 27566190 DOI: 10.1016/j.preteyeres.2016.08.001] [Citation(s) in RCA: 163] [Impact Index Per Article: 18.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2016] [Revised: 08/15/2016] [Accepted: 08/18/2016] [Indexed: 02/06/2023]
Abstract
Autophagy is a catabolic pathway that promotes the degradation and recycling of cellular components. Proteins, lipids, and even whole organelles are engulfed in autophagosomes and delivered to the lysosome for elimination. In response to stress, autophagy mediates the degradation of cell components, which are recycled to generate the nutrients and building blocks required to sustain cellular homeostasis. Moreover, it plays an important role in cellular quality control, particularly in neurons, in which the total burden of altered proteins and damaged organelles cannot be reduced by redistribution to daughter cells through cell division. Research has only begun to examine the role of autophagy in the visual system. The retina, a light-sensitive tissue, detects and transmits electrical impulses through the optic nerve to the visual cortex in the brain. Both the retina and the eye are exposed to a variety of environmental insults and stressors, including genetic mutations and age-associated alterations that impair their function. Here, we review the main studies that have sought to explain autophagy's importance in visual function. We describe the role of autophagy in retinal development and cell differentiation, and discuss the implications of autophagy dysregulation both in physiological aging and in important diseases such as age-associated macular degeneration and glaucoma. We also address the putative role of autophagy in promoting photoreceptor survival and discuss how selective autophagy could provide alternative means of protecting retinal cells. The findings reviewed here underscore the important role of autophagy in maintaining proper retinal function and highlight novel therapeutic approaches for blindness and other diseases of the eye.
Collapse
Affiliation(s)
- Patricia Boya
- Autophagy Lab, Department of Cellular and Molecular Biology, Centro de Investigaciones Biológicas, CSIC, Ramiro de Maeztu 9, 28040, Madrid, Spain.
| | - Lorena Esteban-Martínez
- Autophagy Lab, Department of Cellular and Molecular Biology, Centro de Investigaciones Biológicas, CSIC, Ramiro de Maeztu 9, 28040, Madrid, Spain
| | - Ana Serrano-Puebla
- Autophagy Lab, Department of Cellular and Molecular Biology, Centro de Investigaciones Biológicas, CSIC, Ramiro de Maeztu 9, 28040, Madrid, Spain
| | - Raquel Gómez-Sintes
- Autophagy Lab, Department of Cellular and Molecular Biology, Centro de Investigaciones Biológicas, CSIC, Ramiro de Maeztu 9, 28040, Madrid, Spain
| | - Beatriz Villarejo-Zori
- Autophagy Lab, Department of Cellular and Molecular Biology, Centro de Investigaciones Biológicas, CSIC, Ramiro de Maeztu 9, 28040, Madrid, Spain
| |
Collapse
|