1
|
Kim YA, Choi Y, Kim TG, Jeong J, Yu S, Kim T, Sheen K, Lee Y, Choi T, Park YH, Kang MS, Kim MS. Multi-System-Level Analysis with RNA-Seq on Pterygium Inflammation Discovers Association between Inflammatory Responses, Oxidative Stress, and Oxidative Phosphorylation. Int J Mol Sci 2024; 25:4789. [PMID: 38732006 PMCID: PMC11083828 DOI: 10.3390/ijms25094789] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Revised: 04/18/2024] [Accepted: 04/25/2024] [Indexed: 05/13/2024] Open
Abstract
A pterygium is a common conjunctival degeneration and inflammatory condition. It grows onto the corneal surface or limbus, causing blurred vision and cosmetic issues. Ultraviolet is a well-known risk factor for the development of a pterygium, although its pathogenesis remains unclear, with only limited understanding of its hereditary basis. In this study, we collected RNA-seq from both pterygial tissues and conjunctival tissues (as controls) from six patients (a total of twelve biological samples) and retrieved publicly available data, including eight pterygium samples and eight controls. We investigated the intrinsic gene regulatory mechanisms closely linked to the inflammatory reactions of pterygiums and compared Asian (Korea) and the European (Germany) pterygiums using multiple analysis approaches from different perspectives. The increased expression of antioxidant genes in response to oxidative stress and DNA damage implies an association between these factors and pterygium development. Also, our comparative analysis revealed both similarities and differences between Asian and European pterygiums. The decrease in gene expressions involved in the three primary inflammatory signaling pathways-JAK/STAT, MAPK, and NF-kappa B signaling-suggests a connection between pathway dysfunction and pterygium development. We also observed relatively higher activity of autophagy and antioxidants in the Asian group, while the European group exhibited more pronounced stress responses against oxidative stress. These differences could potentially be necessitated by energy-associated pathways, specifically oxidative phosphorylation.
Collapse
Affiliation(s)
- Ye-Ah Kim
- Translational-Transdisciplinary Research Center, Clinical Research Institute, Kyung Hee University Hospital at Gangdong, Kyung Hee University College of Medicine, Seoul 05278, Republic of Korea; (Y.-A.K.); (Y.C.); (J.J.); (S.Y.); (T.K.); (K.S.); (Y.L.)
- Department of Biomedical Science and Technology, Graduate School, Kyung Hee University, Seoul 02453, Republic of Korea
| | - Yueun Choi
- Translational-Transdisciplinary Research Center, Clinical Research Institute, Kyung Hee University Hospital at Gangdong, Kyung Hee University College of Medicine, Seoul 05278, Republic of Korea; (Y.-A.K.); (Y.C.); (J.J.); (S.Y.); (T.K.); (K.S.); (Y.L.)
- Department of Biomedical Science and Technology, Graduate School, Kyung Hee University, Seoul 02453, Republic of Korea
| | - Tae Gi Kim
- Department of Ophthalmology, Kyung Hee University Hospital at Gangdong, Kyung Hee University College of Medicine, Seoul 05278, Republic of Korea;
| | - Jisu Jeong
- Translational-Transdisciplinary Research Center, Clinical Research Institute, Kyung Hee University Hospital at Gangdong, Kyung Hee University College of Medicine, Seoul 05278, Republic of Korea; (Y.-A.K.); (Y.C.); (J.J.); (S.Y.); (T.K.); (K.S.); (Y.L.)
- Department of Biomedical Science and Technology, Graduate School, Kyung Hee University, Seoul 02453, Republic of Korea
| | - Sanghyeon Yu
- Translational-Transdisciplinary Research Center, Clinical Research Institute, Kyung Hee University Hospital at Gangdong, Kyung Hee University College of Medicine, Seoul 05278, Republic of Korea; (Y.-A.K.); (Y.C.); (J.J.); (S.Y.); (T.K.); (K.S.); (Y.L.)
- Department of Biomedical Science and Technology, Graduate School, Kyung Hee University, Seoul 02453, Republic of Korea
| | - Taeyoon Kim
- Translational-Transdisciplinary Research Center, Clinical Research Institute, Kyung Hee University Hospital at Gangdong, Kyung Hee University College of Medicine, Seoul 05278, Republic of Korea; (Y.-A.K.); (Y.C.); (J.J.); (S.Y.); (T.K.); (K.S.); (Y.L.)
- Department of Biomedical Science and Technology, Graduate School, Kyung Hee University, Seoul 02453, Republic of Korea
| | - Kisung Sheen
- Translational-Transdisciplinary Research Center, Clinical Research Institute, Kyung Hee University Hospital at Gangdong, Kyung Hee University College of Medicine, Seoul 05278, Republic of Korea; (Y.-A.K.); (Y.C.); (J.J.); (S.Y.); (T.K.); (K.S.); (Y.L.)
- Department of Biomedical Science and Technology, Graduate School, Kyung Hee University, Seoul 02453, Republic of Korea
| | - Yoonsung Lee
- Translational-Transdisciplinary Research Center, Clinical Research Institute, Kyung Hee University Hospital at Gangdong, Kyung Hee University College of Medicine, Seoul 05278, Republic of Korea; (Y.-A.K.); (Y.C.); (J.J.); (S.Y.); (T.K.); (K.S.); (Y.L.)
| | - Taesoo Choi
- Department of Urology, School of Medicine, Kyung Hee University, Seoul 05278, Republic of Korea;
| | - Yong Hwan Park
- Department of Microbiology, Ajou University School of Medicine, Suwon 16499, Republic of Korea;
| | - Min Seok Kang
- Department of Ophthalmology, Kyung Hee University Hospital, Kyung Hee University College of Medicine, Seoul 02447, Republic of Korea
| | - Man S. Kim
- Translational-Transdisciplinary Research Center, Clinical Research Institute, Kyung Hee University Hospital at Gangdong, Kyung Hee University College of Medicine, Seoul 05278, Republic of Korea; (Y.-A.K.); (Y.C.); (J.J.); (S.Y.); (T.K.); (K.S.); (Y.L.)
| |
Collapse
|
2
|
Mishra P, Sivakumar A, Johnson A, Pernaci C, Warden AS, El-Hachem LR, Hansen E, Badell-Grau RA, Khare V, Ramirez G, Gillette S, Solis AB, Guo P, Coufal N, Cherqui S. Gene editing improves endoplasmic reticulum-mitochondrial contacts and unfolded protein response in Friedreich's ataxia iPSC-derived neurons. Front Pharmacol 2024; 15:1323491. [PMID: 38420191 PMCID: PMC10899513 DOI: 10.3389/fphar.2024.1323491] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Accepted: 01/16/2024] [Indexed: 03/02/2024] Open
Abstract
Friedreich ataxia (FRDA) is a multisystemic, autosomal recessive disorder caused by homozygous GAA expansion mutation in the first intron of frataxin (FXN) gene. FXN is a mitochondrial protein critical for iron-sulfur cluster biosynthesis and deficiency impairs mitochondrial electron transport chain functions and iron homeostasis within the organelle. Currently, there is no effective treatment for FRDA. We have previously demonstrated that single infusion of wild-type hematopoietic stem and progenitor cells (HSPCs) resulted in prevention of neurologic and cardiac complications of FRDA in YG8R mice, and rescue was mediated by FXN transfer from tissue engrafted, HSPC-derived microglia/macrophages to diseased neurons/myocytes. For a future clinical translation, we developed an autologous stem cell transplantation approach using CRISPR/Cas9 for the excision of the GAA repeats in FRDA patients' CD34+ HSPCs; this strategy leading to increased FXN expression and improved mitochondrial functions. The aim of the current study is to validate the efficiency and safety of our gene editing approach in a disease-relevant model. We generated a cohort of FRDA patient-derived iPSCs and isogenic lines that were gene edited with our CRISPR/Cas9 approach. iPSC derived FRDA neurons displayed characteristic apoptotic and mitochondrial phenotype of the disease, such as non-homogenous microtubule staining in neurites, increased caspase-3 expression, mitochondrial superoxide levels, mitochondrial fragmentation, and partial degradation of the cristae compared to healthy controls. These defects were fully prevented in the gene edited neurons. RNASeq analysis of FRDA and gene edited neurons demonstrated striking improvement in gene clusters associated with endoplasmic reticulum (ER) stress in the isogenic lines. Gene edited neurons demonstrated improved ER-calcium release, normalization of ER stress response gene, XBP-1, and significantly increased ER-mitochondrial contacts that are critical for functional homeostasis of both organelles, as compared to FRDA neurons. Ultrastructural analysis for these contact sites displayed severe ER structural damage in FRDA neurons, that was undetected in gene edited neurons. Taken together, these results represent a novel finding for disease pathogenesis showing dramatic ER structural damage in FRDA, validate the efficacy profile of our FXN gene editing approach in a disease relevant model, and support our approach as an effective strategy for therapeutic intervention for Friedreich's ataxia.
Collapse
Affiliation(s)
- Priyanka Mishra
- Department of Pediatrics, Division of Genetics, University of California, San Diego, San Diego, CA, United States
| | - Anusha Sivakumar
- Department of Pediatrics, Division of Genetics, University of California, San Diego, San Diego, CA, United States
| | - Avalon Johnson
- Department of Pediatrics, University of California, San Diego, San Diego, CA, United States
- Sanford Consortium for Regenerative Medicine, La Jolla, CA, United States
| | - Carla Pernaci
- Department of Pediatrics, University of California, San Diego, San Diego, CA, United States
- Sanford Consortium for Regenerative Medicine, La Jolla, CA, United States
| | - Anna S. Warden
- Department of Pediatrics, University of California, San Diego, San Diego, CA, United States
- Sanford Consortium for Regenerative Medicine, La Jolla, CA, United States
| | - Lilas Rony El-Hachem
- Department of Pediatrics, Division of Genetics, University of California, San Diego, San Diego, CA, United States
| | - Emily Hansen
- Department of Pediatrics, University of California, San Diego, San Diego, CA, United States
- Sanford Consortium for Regenerative Medicine, La Jolla, CA, United States
| | - Rafael A. Badell-Grau
- Department of Pediatrics, Division of Genetics, University of California, San Diego, San Diego, CA, United States
| | - Veenita Khare
- Department of Pediatrics, Division of Genetics, University of California, San Diego, San Diego, CA, United States
| | - Gabriela Ramirez
- Department of Pediatrics, University of California, San Diego, San Diego, CA, United States
- Sanford Consortium for Regenerative Medicine, La Jolla, CA, United States
| | - Sydney Gillette
- Department of Pediatrics, University of California, San Diego, San Diego, CA, United States
- Sanford Consortium for Regenerative Medicine, La Jolla, CA, United States
| | - Angelyn B. Solis
- Department of Pediatrics, Division of Genetics, University of California, San Diego, San Diego, CA, United States
| | - Peng Guo
- Department of Cellular and Molecular Medicine, University of California, San Diego, San Diego, CA, United States
| | - Nicole Coufal
- Department of Pediatrics, University of California, San Diego, San Diego, CA, United States
- Sanford Consortium for Regenerative Medicine, La Jolla, CA, United States
| | - Stephanie Cherqui
- Department of Pediatrics, Division of Genetics, University of California, San Diego, San Diego, CA, United States
| |
Collapse
|
3
|
Zhang SX, Wang JJ, Starr CR, Lee EJ, Park KS, Zhylkibayev A, Medina A, Lin JH, Gorbatyuk M. The endoplasmic reticulum: Homeostasis and crosstalk in retinal health and disease. Prog Retin Eye Res 2024; 98:101231. [PMID: 38092262 PMCID: PMC11056313 DOI: 10.1016/j.preteyeres.2023.101231] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Revised: 12/05/2023] [Accepted: 12/06/2023] [Indexed: 12/19/2023]
Abstract
The endoplasmic reticulum (ER) is the largest intracellular organelle carrying out a broad range of important cellular functions including protein biosynthesis, folding, and trafficking, lipid and sterol biosynthesis, carbohydrate metabolism, and calcium storage and gated release. In addition, the ER makes close contact with multiple intracellular organelles such as mitochondria and the plasma membrane to actively regulate the biogenesis, remodeling, and function of these organelles. Therefore, maintaining a homeostatic and functional ER is critical for the survival and function of cells. This vital process is implemented through well-orchestrated signaling pathways of the unfolded protein response (UPR). The UPR is activated when misfolded or unfolded proteins accumulate in the ER, a condition known as ER stress, and functions to restore ER homeostasis thus promoting cell survival. However, prolonged activation or dysregulation of the UPR can lead to cell death and other detrimental events such as inflammation and oxidative stress; these processes are implicated in the pathogenesis of many human diseases including retinal disorders. In this review manuscript, we discuss the unique features of the ER and ER stress signaling in the retina and retinal neurons and describe recent advances in the research to uncover the role of ER stress signaling in neurodegenerative retinal diseases including age-related macular degeneration, inherited retinal degeneration, achromatopsia and cone diseases, and diabetic retinopathy. In some chapters, we highlight the complex interactions between the ER and other intracellular organelles focusing on mitochondria and illustrate how ER stress signaling regulates common cellular stress pathways such as autophagy. We also touch upon the integrated stress response in retinal degeneration and diabetic retinopathy. Finally, we provide an update on the current development of pharmacological agents targeting the UPR response and discuss some unresolved questions and knowledge gaps to be addressed by future research.
Collapse
Affiliation(s)
- Sarah X Zhang
- Department of Ophthalmology and Ross Eye Institute, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, State University of New York, Buffalo, NY, United States; Department of Biochemistry, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, State University of New York, Buffalo, NY, United States.
| | - Josh J Wang
- Department of Ophthalmology and Ross Eye Institute, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, State University of New York, Buffalo, NY, United States
| | - Christopher R Starr
- Department of Optometry and Vision Science, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Eun-Jin Lee
- Department of Ophthalmology and Byers Eye Institute, Stanford University, Stanford, CA, United States; VA Palo Alto Healthcare System, Palo Alto, CA, United States; Department of Pathology, Stanford University, Stanford, CA, United States
| | - Karen Sophia Park
- Department of Ophthalmology and Ross Eye Institute, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, State University of New York, Buffalo, NY, United States
| | - Assylbek Zhylkibayev
- Department of Optometry and Vision Science, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Andy Medina
- Department of Ophthalmology and Ross Eye Institute, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, State University of New York, Buffalo, NY, United States
| | - Jonathan H Lin
- Department of Ophthalmology and Byers Eye Institute, Stanford University, Stanford, CA, United States; VA Palo Alto Healthcare System, Palo Alto, CA, United States; Department of Pathology, Stanford University, Stanford, CA, United States
| | - Marina Gorbatyuk
- Department of Optometry and Vision Science, University of Alabama at Birmingham, Birmingham, AL, United States
| |
Collapse
|
4
|
Wang J, Wang X, Zhang M, Lang Y, Chen B, Ye Y, Bai Y, Ding S. The activation of spliced X-box binding protein 1 by isorhynchophylline therapy improves diabetic encephalopathy. Cell Biol Toxicol 2023; 39:2587-2613. [PMID: 36695953 DOI: 10.1007/s10565-022-09789-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Accepted: 12/20/2022] [Indexed: 01/26/2023]
Abstract
The primary symptom of diabetic encephalopathy (DE), a kind of central diabetic neuropathy caused by diabetes mellitus (DM), is cognitive impairment. In addition, the tetracyclic oxindole alkaloid isorhynchophylline (IRN) helps lessen cognitive impairment. However, it is still unclear how IRN affects DM and DE and what mechanisms are involved. The effectiveness of IRN on brain insulin resistance was carefully examined in this work, both in vitro and in vivo. We found that IRN accelerates spliced form of X-box binding protein 1 (sXBP1) translocation into the nucleus under high glucose conditions in vitro. IRN also facilitates the nuclear association of pCREB with sXBP1 and the binding of regulatory subunits of phosphatidylinositol 3-kinase (PI3K) p85α or p85β with XBP1 to restore high glucose impairment. Also, IRN treatment improves high glucose-mediated impairment of insulin signaling, endoplasmic reticulum stress, and pyroptosis/apoptosis by depending on sXBP1 in vitro. In vivo studies suggested that IRN attenuates cognitive impairment, ameliorating peripheral insulin resistance, activating insulin signaling, inactivating activating transcription factor 6 (ATF6) and C/EBP homology protein (CHOP), and mitigating pyroptosis/apoptosis by stimulation of sXBP1 nuclear translocation in the brain. In summary, these data indicate that IRN contributes to maintaining insulin homeostasis by activating sXBP1 in the brain. Thus, IRN is a potent antidiabetic agent as well as an sXBP1 activator that has promising potential for the prevention or treatment of DE.
Collapse
Affiliation(s)
- Jian Wang
- Department of Laboratory Animal Science, Fudan University, Shanghai, 200032, China
- Key Laboratory of Diagnosis and Treatment of Severe Hepato-Pancreatic Diseases of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China
- Huangshi Love & Health Hospital, Hubei Polytechnic University, Huangshi, 435000, China
| | - Xuebao Wang
- Department of Laboratory Animal Science, Fudan University, Shanghai, 200032, China
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325000, Zhejiang, China
| | - Minxue Zhang
- Key Laboratory of Diagnosis and Treatment of Severe Hepato-Pancreatic Diseases of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China
| | - Yan Lang
- Key Laboratory of Diagnosis and Treatment of Severe Hepato-Pancreatic Diseases of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China
| | - Baihui Chen
- Key Laboratory of Diagnosis and Treatment of Severe Hepato-Pancreatic Diseases of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China
| | - Yiru Ye
- School of Information and Engineering, Wenzhou Medical University, Wenzhou, 325035, Zhejiang, China
| | - Yongheng Bai
- Key Laboratory of Diagnosis and Treatment of Severe Hepato-Pancreatic Diseases of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China
| | - Saidan Ding
- Department of Laboratory Animal Science, Fudan University, Shanghai, 200032, China.
- Key Laboratory of Diagnosis and Treatment of Severe Hepato-Pancreatic Diseases of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China.
| |
Collapse
|
5
|
McLaughlin T, Wang G, Medina A, Perkins J, Nihlawi R, Seyfried D, Hu Z, Wang JJ, Zhang SX. Essential Role of XBP1 in Maintaining Photoreceptor Synaptic Integrity in Early Diabetic Retinopathy. Invest Ophthalmol Vis Sci 2023; 64:40. [PMID: 38015176 PMCID: PMC10691399 DOI: 10.1167/iovs.64.14.40] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2023] [Accepted: 11/04/2023] [Indexed: 11/29/2023] Open
Abstract
Purpose Diabetic retinopathy (DR) is a leading cause of blindness in working-age adults characterized by retinal dysfunction and neurovascular degeneration. We previously reported that deletion of X-box binding protein 1 (XBP1) leads to accelerated retinal neurodegeneration in diabetes; however, the mechanisms remain elusive. The goal of this study is to determine the role of XBP1 in the regulation of photoreceptor synaptic integrity in early DR. Methods Diabetes was induced by streptozotocin in retina-specific XBP1 conditional knockout (cKO) or wild-type (WT) mice to generate diabetic cKO (cKO/DM) or WT/DM mice for comparison with nondiabetic cKO (cKO/NDM) and WT/NDM mice. Retinal morphology, structure, and function were assessed by immunohistochemistry, optical coherence tomography, and electroretinogram (ERG) after 3 months of diabetes. The synapses between photoreceptors and bipolar cells were examined by confocal microscopy, and synaptic integrity was quantified using the QUANTOS algorithm. Results We found a thinning of the outer nuclear layer and a decline in the b-wave amplitude in dark- and light-adapted ERG in cKO/DM mice compared to all other groups. In line with these changes, cKO mice showed increased loss of synaptic integrity compared to WT mice, regardless of diabetes status. In searching for candidate molecules responsible for the loss of photoreceptor synaptic integrity in diabetic and XBP1-deficient retinas, we found decreased mRNA and protein levels of DLG4/PSD-95 in cKO/DM retina compared to WT/DM. Conclusions These findings suggest that XBP1 is a crucial regulator in maintaining synaptic integrity and retinal function, possibly through regulation of synaptic scaffold proteins.
Collapse
Affiliation(s)
- Todd McLaughlin
- Department of Ophthalmology and Ross Eye Institute, University at Buffalo, State University of New York, Buffalo, New York, United States
| | - Grant Wang
- Department of Ophthalmology and Ross Eye Institute, University at Buffalo, State University of New York, Buffalo, New York, United States
| | - Andy Medina
- Department of Ophthalmology and Ross Eye Institute, University at Buffalo, State University of New York, Buffalo, New York, United States
| | - Jacob Perkins
- Department of Ophthalmology and Ross Eye Institute, University at Buffalo, State University of New York, Buffalo, New York, United States
| | - Rhudwan Nihlawi
- Department of Ophthalmology and Ross Eye Institute, University at Buffalo, State University of New York, Buffalo, New York, United States
| | - Don Seyfried
- Department of Ophthalmology and Ross Eye Institute, University at Buffalo, State University of New York, Buffalo, New York, United States
| | - Zihua Hu
- Department of Ophthalmology and Ross Eye Institute, University at Buffalo, State University of New York, Buffalo, New York, United States
- Center for Computational Research, New York State Center of Excellence in Bioinformatics and Life Sciences, State University of New York, Buffalo, New York, United States
| | - Joshua J. Wang
- Department of Ophthalmology and Ross Eye Institute, University at Buffalo, State University of New York, Buffalo, New York, United States
- Department of Biochemistry, State University of New York, Buffalo, New York, United States
| | - Sarah X. Zhang
- Department of Ophthalmology and Ross Eye Institute, University at Buffalo, State University of New York, Buffalo, New York, United States
- Department of Biochemistry, State University of New York, Buffalo, New York, United States
| |
Collapse
|
6
|
Chen X, Shi C, He M, Xiong S, Xia X. Endoplasmic reticulum stress: molecular mechanism and therapeutic targets. Signal Transduct Target Ther 2023; 8:352. [PMID: 37709773 PMCID: PMC10502142 DOI: 10.1038/s41392-023-01570-w] [Citation(s) in RCA: 88] [Impact Index Per Article: 88.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Revised: 06/17/2023] [Accepted: 07/14/2023] [Indexed: 09/16/2023] Open
Abstract
The endoplasmic reticulum (ER) functions as a quality-control organelle for protein homeostasis, or "proteostasis". The protein quality control systems involve ER-associated degradation, protein chaperons, and autophagy. ER stress is activated when proteostasis is broken with an accumulation of misfolded and unfolded proteins in the ER. ER stress activates an adaptive unfolded protein response to restore proteostasis by initiating protein kinase R-like ER kinase, activating transcription factor 6, and inositol requiring enzyme 1. ER stress is multifaceted, and acts on aspects at the epigenetic level, including transcription and protein processing. Accumulated data indicates its key role in protein homeostasis and other diverse functions involved in various ocular diseases, such as glaucoma, diabetic retinopathy, age-related macular degeneration, retinitis pigmentosa, achromatopsia, cataracts, ocular tumors, ocular surface diseases, and myopia. This review summarizes the molecular mechanisms underlying the aforementioned ocular diseases from an ER stress perspective. Drugs (chemicals, neurotrophic factors, and nanoparticles), gene therapy, and stem cell therapy are used to treat ocular diseases by alleviating ER stress. We delineate the advancement of therapy targeting ER stress to provide new treatment strategies for ocular diseases.
Collapse
Affiliation(s)
- Xingyi Chen
- Eye Center of Xiangya Hospital, Central South University, 410008, Changsha, Hunan, China
- Hunan Key Laboratory of Ophthalmology, Central South University, 410008, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Chaoran Shi
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Meihui He
- Eye Center of Xiangya Hospital, Central South University, 410008, Changsha, Hunan, China
- Hunan Key Laboratory of Ophthalmology, Central South University, 410008, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Siqi Xiong
- Eye Center of Xiangya Hospital, Central South University, 410008, Changsha, Hunan, China.
- Hunan Key Laboratory of Ophthalmology, Central South University, 410008, Changsha, China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China.
| | - Xiaobo Xia
- Eye Center of Xiangya Hospital, Central South University, 410008, Changsha, Hunan, China.
- Hunan Key Laboratory of Ophthalmology, Central South University, 410008, Changsha, China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China.
| |
Collapse
|
7
|
McLaughlin T, Wang J, Jia L, Wu F, Wang Y, Wang JJ, Mu X, Zhang SX. Neuronal p58 IPK Protects Retinal Ganglion Cells Independently of Macrophage/Microglia Activation in Ocular Hypertension. Cells 2023; 12:1558. [PMID: 37371028 PMCID: PMC10297187 DOI: 10.3390/cells12121558] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Revised: 01/10/2023] [Accepted: 01/12/2023] [Indexed: 06/29/2023] Open
Abstract
p58IPK is a multifaceted endoplasmic reticulum (ER) chaperone and a regulator of eIF2α kinases involved in a wide range of cellular processes including protein synthesis, ER stress response, and macrophage-mediated inflammation. Systemic deletion of p58IPK leads to age-related loss of retinal ganglion cells (RGC) and exacerbates RGC damage induced by ischemia/reperfusion and increased intraocular pressure (IOP), suggesting a protective role of p58IPK in the retina. However, the mechanisms remain elusive. Herein, we investigated the cellular mechanisms underlying the neuroprotection action of p58IPK using conditional knockout (cKO) mouse lines where p58IPK is deleted in retinal neurons (Chx10-p58IPK cKO) or in myeloid cells (Lyz2-p58IPK cKO). In addition, we overexpressed p58IPK by adeno-associated virus (AAV) in the retina to examine the effect of p58IPK on RGC survival after ocular hypertension (OHT) in wild type (WT) mice. Our results show that overexpression of p58IPK by AAV significantly improved RGC survival after OHT in WT mice, suggesting a protective effect of p58IPK on reducing RGC injury. Conditional knockout of p58IPK in retinal neurons or in myeloid cells did not alter retinal structure or cellular composition. However, a significant reduction in the b wave of light-adapted electroretinogram (ERG) was observed in Chx10-p58IPK cKO mice. Deletion of p58IPK in retinal neurons exacerbates RGC loss at 14 days after OHT. In contrast, deficiency of p58IPK in myeloid cells increased the microglia/macrophage activation but had no effect on RGC loss. We conclude that deletion of p58IPK in macrophages increases their activation, but does not influence RGC survival. These results suggest that the neuroprotective action of p58IPK is mediated by its expression in retinal neurons, but not in macrophages. Therefore, targeting p58IPK specifically in retinal neurons is a promising approach for the treatment of neurodegenerative retinal diseases including glaucoma.
Collapse
Affiliation(s)
- Todd McLaughlin
- Department of Ophthalmology and Ross Eye Institute, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, State University of New York, Buffalo, NY 14203, USA
| | - Jinli Wang
- Department of Ophthalmology and Ross Eye Institute, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, State University of New York, Buffalo, NY 14203, USA
| | - Liyun Jia
- Department of Ophthalmology and Ross Eye Institute, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, State University of New York, Buffalo, NY 14203, USA
- Beijing Tongren Hospital, Capital Medical University, Beijing 100005, China
| | - Fuguo Wu
- Department of Ophthalmology and Ross Eye Institute, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, State University of New York, Buffalo, NY 14203, USA
| | - Yaqin Wang
- Department of Ophthalmology and Ross Eye Institute, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, State University of New York, Buffalo, NY 14203, USA
- Taihe Hospital, Hubei University of Medicine, Shiyan 442005, China
| | - Joshua J. Wang
- Department of Ophthalmology and Ross Eye Institute, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, State University of New York, Buffalo, NY 14203, USA
| | - Xiuqian Mu
- Department of Ophthalmology and Ross Eye Institute, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, State University of New York, Buffalo, NY 14203, USA
- Department of Biochemistry, University at Buffalo, State University of New York, Buffalo, NY 14203, USA
| | - Sarah X. Zhang
- Department of Ophthalmology and Ross Eye Institute, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, State University of New York, Buffalo, NY 14203, USA
- Department of Biochemistry, University at Buffalo, State University of New York, Buffalo, NY 14203, USA
| |
Collapse
|
8
|
Massoudi D, Gorman S, Kuo YM, Iwawaki T, Oakes SA, Papa FR, Gould DB. Deletion of the Unfolded Protein Response Transducer IRE1α Is Detrimental to Aging Photoreceptors and to ER Stress-Mediated Retinal Degeneration. Invest Ophthalmol Vis Sci 2023; 64:30. [PMID: 37097227 PMCID: PMC10148664 DOI: 10.1167/iovs.64.4.30] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Accepted: 04/06/2023] [Indexed: 04/26/2023] Open
Abstract
Purpose The unfolded protein response (UPR) is triggered when the protein folding capacity of the endoplasmic reticulum (ER) is overwhelmed and misfolded proteins accumulate in the ER, a condition referred to as ER stress. IRE1α is an ER-resident protein that plays major roles in orchestrating the UPR. Several lines of evidence implicate the UPR and its transducers in neurodegenerative diseases, including retinitis pigmentosa (RP), a group of inherited diseases that cause progressive dysfunction and loss of rod and cone photoreceptors. This study evaluated the contribution of IRE1α to photoreceptor development, homeostasis, and degeneration. Methods We used a conditional gene targeting strategy to selectively inactivate Ire1α in mouse rod photoreceptors. We used a combination of optical coherence tomography (OCT) imaging, histology, and electroretinography (ERG) to assess longitudinally the effect of IRE1α deficiency in retinal development and function. Furthermore, we evaluated the IRE1α-deficient retina responses to tunicamycin-induced ER stress and in the context of RP caused by the rhodopsin mutation RhoP23H. Results OCT imaging, histology, and ERG analyses did not reveal abnormalities in IRE1α-deficient retinas up to 3 months old. However, by 6 months of age, the Ire1α mutant animals showed reduced outer nuclear layer thickness and deficits in retinal function. Furthermore, conditional inactivation of Ire1α in rod photoreceptors accelerated retinal degeneration caused by the RhoP23H mutation. Conclusions These data suggest that IRE1α is dispensable for photoreceptor development but important for photoreceptor homeostasis in aging retinas and for protecting against ER stress-mediated photoreceptor degeneration.
Collapse
Affiliation(s)
- Dawiyat Massoudi
- Department of Ophthalmology, University of California, San Francisco, San Francisco, California, United States
| | - Seán Gorman
- Department of Ophthalmology, University of California, San Francisco, San Francisco, California, United States
| | - Yien-Ming Kuo
- Department of Ophthalmology, University of California, San Francisco, San Francisco, California, United States
| | - Takao Iwawaki
- Division of Cell Medicine, Medical Research Institute, Kanazawa Medical University, Ishikawa, Japan
| | - Scott A. Oakes
- Department of Pathology, Pritzker School of Medicine, University of Chicago, Chicago, Illinois, United States
| | - Feroz R. Papa
- Department of Medicine, Diabetes Center, Quantitative Biosciences Institute and Lung Biology Center University of California, San Francisco, San Francisco, California, United States
| | - Douglas B. Gould
- Department of Ophthalmology, University of California, San Francisco, San Francisco, California, United States
- Department of Anatomy, Institute for Human Genetics, Cardiovascular Research Institute, Bakar Aging Research Institute, University of California, San Francisco, California, United States
| |
Collapse
|
9
|
Angueyra JM, Kunze VP, Patak LK, Kim H, Kindt K, Li W. Transcription factors underlying photoreceptor diversity. eLife 2023; 12:e81579. [PMID: 36745553 PMCID: PMC9901936 DOI: 10.7554/elife.81579] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2022] [Accepted: 01/22/2023] [Indexed: 02/07/2023] Open
Abstract
During development, retinal progenitors navigate a complex landscape of fate decisions to generate the major cell classes necessary for proper vision. Transcriptional regulation is critical to generate diversity within these major cell classes. Here, we aim to provide the resources and techniques required to identify transcription factors necessary to generate and maintain diversity in photoreceptor subtypes, which are critical for vision. First, we generate a key resource: a high-quality and deep transcriptomic profile of each photoreceptor subtype in adult zebrafish. We make this resource openly accessible, easy to explore, and have integrated it with other currently available photoreceptor transcriptomic datasets. Second, using our transcriptomic profiles, we derive an in-depth map of expression of transcription factors in photoreceptors. Third, we use efficient CRISPR-Cas9 based mutagenesis to screen for null phenotypes in F0 larvae (F0 screening) as a fast, efficient, and versatile technique to assess the involvement of candidate transcription factors in the generation of photoreceptor subtypes. We first show that known phenotypes can be easily replicated using this method: loss of S cones in foxq2 mutants and loss of rods in nr2e3 mutants. We then identify novel functions for the transcription factor Tbx2, demonstrating that it plays distinct roles in controlling the generation of all photoreceptor subtypes within the retina. Our study provides a roadmap to discover additional factors involved in this process. Additionally, we explore four transcription factors of unknown function (Skor1a, Sall1a, Lrrfip1a, and Xbp1), and find no evidence for their involvement in the generation of photoreceptor subtypes. This dataset and screening method will be a valuable way to explore the genes involved in many other essential aspects of photoreceptor biology.
Collapse
Affiliation(s)
- Juan M Angueyra
- Unit of Retinal Neurophysiology, National Eye Institute, National Institutes of HealthBethesdaUnited States
| | - Vincent P Kunze
- Unit of Retinal Neurophysiology, National Eye Institute, National Institutes of HealthBethesdaUnited States
| | - Laura K Patak
- Unit of Retinal Neurophysiology, National Eye Institute, National Institutes of HealthBethesdaUnited States
| | - Hailey Kim
- Unit of Retinal Neurophysiology, National Eye Institute, National Institutes of HealthBethesdaUnited States
| | - Katie Kindt
- Section on Sensory Cell Development and Function, National Institute on Deafness and Other Communication Disorders, National Institutes of HealthBethesdaUnited States
| | - Wei Li
- Unit of Retinal Neurophysiology, National Eye Institute, National Institutes of HealthBethesdaUnited States
| |
Collapse
|
10
|
Enhanced IRE1α Phosphorylation/Oligomerization-Triggered XBP1 Splicing Contributes to Parkin-Mediated Prevention of SH-SY5Y Cell Death under Nitrosative Stress. Int J Mol Sci 2023; 24:ijms24032017. [PMID: 36768338 PMCID: PMC9917145 DOI: 10.3390/ijms24032017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Revised: 01/10/2023] [Accepted: 01/17/2023] [Indexed: 01/21/2023] Open
Abstract
Mutations in parkin, a neuroprotective protein, are the predominant cause of autosomal recessive juvenile Parkinson's disease. Neuroinflammation-derived nitrosative stress has been implicated in the etiology of the chronic neurodegeneration. However, the interactions between genetic predisposition and nitrosative stress contributing to the degeneration of dopaminergic (DA) neurons remain incompletely understood. Here, we used the SH-SY5Y neuroblastoma cells to investigate the function of parkin and its pathogenic mutants in relation to cell survival under nitric oxide (NO) exposure. The results showed that overexpression of wild-type parkin protected SH-SY5Y cells from NO-induced apoptosis in a reactive oxygen species-dependent manner. Under nitrosative stress conditions, parkin selectively upregulated the inositol-requiring enzyme 1α/X-box binding protein 1 (IRE1α/XBP1) signaling axis, an unfolded protein response signal through the sensor IRE1α, which controls the splicing of XBP1 mRNA. Inhibition of XBP1 mRNA splicing either by pharmacologically inhibiting IRE1α endoribonuclease activity or by genetically knocking down XBP1 interfered with the protective activity of parkin. Furthermore, pathogenic parkin mutants with a defective protective capacity showed a lower ability to activate the IRE1α/XBP1 signaling. Finally, we demonstrated that IRE1α activity augmented by parkin was possibly mediated through interacting with IRE1α to regulate its phosphorylation/oligomerization processes, whereas mutant parkin diminished its binding to and activation of IRE1α. Thus, these results support a direct link between the protective activity of parkin and the IRE1α/XBP1 pathway in response to nitrosative stress, and mutant parkin disrupts this function.
Collapse
|
11
|
Tang X, Wang J, Abboud HE, Chen Y, Wang JJ, Zhang SX. Sustained Upregulation of Endothelial Nox4 Mediates Retinal Vascular Pathology in Type 1 Diabetes. Diabetes 2023; 72:112-125. [PMID: 36321974 PMCID: PMC9797318 DOI: 10.2337/db22-0194] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/23/2022] [Accepted: 10/10/2022] [Indexed: 12/24/2022]
Abstract
NADPH oxidase 4 (Nox4) is a major source of reactive oxygen species (ROS) in retinal endothelial cells (ECs) and is upregulated under hyperglycemic and hypoxic conditions. However, the role of endothelial Nox4 upregulation in long-term retinal blood vessel damage in diabetic retinopathy (DR) remains undefined. Here, we attempted to address this question using humanized EC-specific Nox4 transgenic (hNox4EC-Tg) and EC-specific Nox4 knockout (Nox4EC-KO) mouse models. Our results show that hNox4EC-Tg mice at age of 10-12 months exhibited increased tortuosity of retinal blood vessels, focal vascular leakage, and acellular capillary formation. In vitro study revealed enhanced apoptosis in brain microvascular ECs derived from hNox4EC-Tg mice, concomitant with increased mitochondrial ROS, elevated lipid peroxidation, decreased mitochondrial membrane potential, and reduced mitochondrial respiratory function. In contrast, EC-specific deletion of Nox4 decreased mitochondrial ROS generation, alleviated mitochondrial damage, reduced EC apoptosis, and protected the retina from acellular capillary formation and vascular hyperpermeability in a streptozotocin-induced diabetes mouse model. These findings suggest that sustained upregulation of Nox4 in the endothelium contributes to retinal vascular pathology in diabetes, at least in part, through impairing mitochondrial function. Normalization of Nox4 expression in ECs may provide a new approach for prevention of vascular injury in DR.
Collapse
Affiliation(s)
- Xixiang Tang
- Department of Ophthalmology and Ross Eye Institute, University at Buffalo, State University of New York, Buffalo, NY
- SUNY Eye Institute, State University of New York, Buffalo, NY
- Department of Endocrinology and Metabolism, Third Affiliated Hospital, Sun Yat-sen University; Guangdong Provincial Key Laboratory of Diabetology, Guangzhou, China
- VIP Medical Service Center, Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Jinli Wang
- Department of Ophthalmology and Ross Eye Institute, University at Buffalo, State University of New York, Buffalo, NY
- SUNY Eye Institute, State University of New York, Buffalo, NY
| | - Hanna E. Abboud
- Department of Medicine, South Texas Veterans Healthcare System and the University of Texas Health Science Center, San Antonio, TX
| | - Yanming Chen
- Department of Endocrinology and Metabolism, Third Affiliated Hospital, Sun Yat-sen University; Guangdong Provincial Key Laboratory of Diabetology, Guangzhou, China
| | - Joshua J. Wang
- Department of Ophthalmology and Ross Eye Institute, University at Buffalo, State University of New York, Buffalo, NY
- SUNY Eye Institute, State University of New York, Buffalo, NY
| | - Sarah X. Zhang
- Department of Ophthalmology and Ross Eye Institute, University at Buffalo, State University of New York, Buffalo, NY
- SUNY Eye Institute, State University of New York, Buffalo, NY
- Department of Biochemistry, University at Buffalo, State University of New York, Buffalo, NY
| |
Collapse
|
12
|
Guo J, Huang X, Dou L, Yan M, Shen T, Tang W, Li J. Aging and aging-related diseases: from molecular mechanisms to interventions and treatments. Signal Transduct Target Ther 2022; 7:391. [PMID: 36522308 PMCID: PMC9755275 DOI: 10.1038/s41392-022-01251-0] [Citation(s) in RCA: 305] [Impact Index Per Article: 152.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Revised: 11/03/2022] [Accepted: 11/10/2022] [Indexed: 12/23/2022] Open
Abstract
Aging is a gradual and irreversible pathophysiological process. It presents with declines in tissue and cell functions and significant increases in the risks of various aging-related diseases, including neurodegenerative diseases, cardiovascular diseases, metabolic diseases, musculoskeletal diseases, and immune system diseases. Although the development of modern medicine has promoted human health and greatly extended life expectancy, with the aging of society, a variety of chronic diseases have gradually become the most important causes of disability and death in elderly individuals. Current research on aging focuses on elucidating how various endogenous and exogenous stresses (such as genomic instability, telomere dysfunction, epigenetic alterations, loss of proteostasis, compromise of autophagy, mitochondrial dysfunction, cellular senescence, stem cell exhaustion, altered intercellular communication, deregulated nutrient sensing) participate in the regulation of aging. Furthermore, thorough research on the pathogenesis of aging to identify interventions that promote health and longevity (such as caloric restriction, microbiota transplantation, and nutritional intervention) and clinical treatment methods for aging-related diseases (depletion of senescent cells, stem cell therapy, antioxidative and anti-inflammatory treatments, and hormone replacement therapy) could decrease the incidence and development of aging-related diseases and in turn promote healthy aging and longevity.
Collapse
Affiliation(s)
- Jun Guo
- The Key Laboratory of Geriatrics, Beijing Institute of Geriatrics, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital/National Center of Gerontology of National Health Commission, Beijing, 100730, China
| | - Xiuqing Huang
- The Key Laboratory of Geriatrics, Beijing Institute of Geriatrics, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital/National Center of Gerontology of National Health Commission, Beijing, 100730, China
| | - Lin Dou
- The Key Laboratory of Geriatrics, Beijing Institute of Geriatrics, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital/National Center of Gerontology of National Health Commission, Beijing, 100730, China
| | - Mingjing Yan
- The Key Laboratory of Geriatrics, Beijing Institute of Geriatrics, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital/National Center of Gerontology of National Health Commission, Beijing, 100730, China
| | - Tao Shen
- The Key Laboratory of Geriatrics, Beijing Institute of Geriatrics, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital/National Center of Gerontology of National Health Commission, Beijing, 100730, China.
| | - Weiqing Tang
- The Key Laboratory of Geriatrics, Beijing Institute of Geriatrics, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital/National Center of Gerontology of National Health Commission, Beijing, 100730, China.
| | - Jian Li
- The Key Laboratory of Geriatrics, Beijing Institute of Geriatrics, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital/National Center of Gerontology of National Health Commission, Beijing, 100730, China.
| |
Collapse
|
13
|
Cai Y, Song W, Li J, Jing Y, Liang C, Zhang L, Zhang X, Zhang W, Liu B, An Y, Li J, Tang B, Pei S, Wu X, Liu Y, Zhuang CL, Ying Y, Dou X, Chen Y, Xiao FH, Li D, Yang R, Zhao Y, Wang Y, Wang L, Li Y, Ma S, Wang S, Song X, Ren J, Zhang L, Wang J, Zhang W, Xie Z, Qu J, Wang J, Xiao Y, Tian Y, Wang G, Hu P, Ye J, Sun Y, Mao Z, Kong QP, Liu Q, Zou W, Tian XL, Xiao ZX, Liu Y, Liu JP, Song M, Han JDJ, Liu GH. The landscape of aging. SCIENCE CHINA. LIFE SCIENCES 2022; 65:2354-2454. [PMID: 36066811 PMCID: PMC9446657 DOI: 10.1007/s11427-022-2161-3] [Citation(s) in RCA: 125] [Impact Index Per Article: 62.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Accepted: 07/05/2022] [Indexed: 02/07/2023]
Abstract
Aging is characterized by a progressive deterioration of physiological integrity, leading to impaired functional ability and ultimately increased susceptibility to death. It is a major risk factor for chronic human diseases, including cardiovascular disease, diabetes, neurological degeneration, and cancer. Therefore, the growing emphasis on "healthy aging" raises a series of important questions in life and social sciences. In recent years, there has been unprecedented progress in aging research, particularly the discovery that the rate of aging is at least partly controlled by evolutionarily conserved genetic pathways and biological processes. In an attempt to bring full-fledged understanding to both the aging process and age-associated diseases, we review the descriptive, conceptual, and interventive aspects of the landscape of aging composed of a number of layers at the cellular, tissue, organ, organ system, and organismal levels.
Collapse
Affiliation(s)
- Yusheng Cai
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, 100101, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, 100101, China
| | - Wei Song
- Frontier Science Center for Immunology and Metabolism, Medical Research Institute, College of Life Sciences, Wuhan University, Wuhan, 430071, China
| | - Jiaming Li
- CAS Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences and China National Center for Bioinformation, Beijing, 100101, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Ying Jing
- University of Chinese Academy of Sciences, Beijing, 100049, China
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Chuqian Liang
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, 100101, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, 100101, China
| | - Liyuan Zhang
- CAS Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences and China National Center for Bioinformation, Beijing, 100101, China
| | - Xia Zhang
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Wenhui Zhang
- University of Chinese Academy of Sciences, Beijing, 100049, China
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Beibei Liu
- CAS Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences and China National Center for Bioinformation, Beijing, 100101, China
| | - Yongpan An
- Peking University International Cancer Institute, Peking University Health Science Center, Peking University, Beijing, 100191, China
| | - Jingyi Li
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, 100101, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, 100101, China
| | - Baixue Tang
- School of Pharmaceutical Sciences, Tsinghua University, Beijing, 100084, China
| | - Siyu Pei
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Xueying Wu
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Yuxuan Liu
- School of Pharmaceutical Sciences, Beijing Advanced Innovation Center for Structural Biology, Ministry of Education Key Laboratory of Bioorganic Phosphorus Chemistry and Chemical Biology, Tsinghua University, Beijing, 100084, China
| | - Cheng-Le Zhuang
- Colorectal Cancer Center/Department of Gastrointestinal Surgery, Shanghai Tenth People's Hospital Affiliated to Tongji University, Shanghai, 200072, China
| | - Yilin Ying
- Department of Geriatrics, Medical Center on Aging of Shanghai Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200025, China
- International Laboratory in Hematology and Cancer, Shanghai Jiaotong University School of Medicine/Ruijin Hospital, Shanghai, 200025, China
| | - Xuefeng Dou
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Yu Chen
- Shanghai Key Laboratory of Maternal Fetal Medicine, Clinical and Translational Research Center of Shanghai First Maternity and Infant Hospital, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China
| | - Fu-Hui Xiao
- State Key Laboratory of Genetic Resources and Evolution/Key Laboratory of Healthy Aging Research of Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650223, China
- CAS Center for Excellence in Animal Evolution and Genetics, Chinese Academy of Sciences, Kunming, 650223, China
| | - Dingfeng Li
- Institute on Aging and Brain Disorders, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230027, China
| | - Ruici Yang
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, CAS Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, 200031, China
| | - Ya Zhao
- Aging and Vascular Diseases, Human Aging Research Institute (HARI) and School of Life Science, Nanchang University, and Jiangxi Key Laboratory of Human Aging, Nanchang, 330031, China
| | - Yang Wang
- Center of Growth, Metabolism and Aging, Key Laboratory of Bio-Resource and Eco-Environment, Ministry of Education, College of Life Sciences, Sichuan University, Chengdu, 610065, China
| | - Lihui Wang
- Institute of Ageing Research, Hangzhou Normal University, School of Basic Medical Sciences, Hangzhou, 311121, China
| | - Yujing Li
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, 100101, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, 100101, China
| | - Shuai Ma
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China.
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, 100101, China.
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, 100101, China.
| | - Si Wang
- Advanced Innovation Center for Human Brain Protection, National Clinical Research Center for Geriatric Disorders, Xuanwu Hospital Capital Medical University, Beijing, 100053, China.
- Aging Translational Medicine Center, International Center for Aging and Cancer, Beijing Municipal Geriatric Medical Research Center, Xuanwu Hospital Capital Medical University, Beijing, 100053, China.
- The Fifth People's Hospital of Chongqing, Chongqing, 400062, China.
| | - Xiaoyuan Song
- MOE Key Laboratory of Cellular Dynamics, Hefei National Research Center for Physical Sciences at the Microscale, CAS Key Laboratory of Brain Function and Disease, Neurodegenerative Disorder Research Center, School of Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230026, China.
| | - Jie Ren
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, 100101, China.
- CAS Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences and China National Center for Bioinformation, Beijing, 100101, China.
- University of Chinese Academy of Sciences, Beijing, 100049, China.
| | - Liang Zhang
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, 100101, China.
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, Shanghai, 200031, China.
| | - Jun Wang
- University of Chinese Academy of Sciences, Beijing, 100049, China.
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, 100101, China.
| | - Weiqi Zhang
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, 100101, China.
- CAS Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences and China National Center for Bioinformation, Beijing, 100101, China.
- University of Chinese Academy of Sciences, Beijing, 100049, China.
- Aging Translational Medicine Center, International Center for Aging and Cancer, Beijing Municipal Geriatric Medical Research Center, Xuanwu Hospital Capital Medical University, Beijing, 100053, China.
| | - Zhengwei Xie
- Peking University International Cancer Institute, Peking University Health Science Center, Peking University, Beijing, 100191, China.
| | - Jing Qu
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, 100101, China.
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, 100101, China.
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China.
| | - Jianwei Wang
- School of Pharmaceutical Sciences, Tsinghua University, Beijing, 100084, China.
| | - Yichuan Xiao
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, Shanghai, 200031, China.
| | - Ye Tian
- University of Chinese Academy of Sciences, Beijing, 100049, China.
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, 100101, China.
| | - Gelin Wang
- School of Pharmaceutical Sciences, Beijing Advanced Innovation Center for Structural Biology, Ministry of Education Key Laboratory of Bioorganic Phosphorus Chemistry and Chemical Biology, Tsinghua University, Beijing, 100084, China.
| | - Ping Hu
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, 100101, China.
- Colorectal Cancer Center/Department of Gastrointestinal Surgery, Shanghai Tenth People's Hospital Affiliated to Tongji University, Shanghai, 200072, China.
- Guangzhou Laboratory, Guangzhou International Bio Island, Guangzhou, 510005, China.
| | - Jing Ye
- Department of Geriatrics, Medical Center on Aging of Shanghai Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200025, China.
- International Laboratory in Hematology and Cancer, Shanghai Jiaotong University School of Medicine/Ruijin Hospital, Shanghai, 200025, China.
| | - Yu Sun
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, Shanghai, 200031, China.
- Department of Medicine and VAPSHCS, University of Washington, Seattle, 98195, USA.
| | - Zhiyong Mao
- Shanghai Key Laboratory of Maternal Fetal Medicine, Clinical and Translational Research Center of Shanghai First Maternity and Infant Hospital, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China.
| | - Qing-Peng Kong
- State Key Laboratory of Genetic Resources and Evolution/Key Laboratory of Healthy Aging Research of Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650223, China.
- CAS Center for Excellence in Animal Evolution and Genetics, Chinese Academy of Sciences, Kunming, 650223, China.
| | - Qiang Liu
- CAS Center for Excellence in Animal Evolution and Genetics, Chinese Academy of Sciences, Kunming, 650223, China.
- Institute on Aging and Brain Disorders, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230027, China.
| | - Weiguo Zou
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, CAS Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, 200031, China.
| | - Xiao-Li Tian
- Aging and Vascular Diseases, Human Aging Research Institute (HARI) and School of Life Science, Nanchang University, and Jiangxi Key Laboratory of Human Aging, Nanchang, 330031, China.
| | - Zhi-Xiong Xiao
- Center of Growth, Metabolism and Aging, Key Laboratory of Bio-Resource and Eco-Environment, Ministry of Education, College of Life Sciences, Sichuan University, Chengdu, 610065, China.
| | - Yong Liu
- Frontier Science Center for Immunology and Metabolism, Medical Research Institute, College of Life Sciences, Wuhan University, Wuhan, 430071, China.
| | - Jun-Ping Liu
- Institute of Ageing Research, Hangzhou Normal University, School of Basic Medical Sciences, Hangzhou, 311121, China.
- Department of Immunology and Pathology, Monash University Faculty of Medicine, Prahran, Victoria, 3181, Australia.
- Hudson Institute of Medical Research, and Monash University Department of Molecular and Translational Science, Clayton, Victoria, 3168, Australia.
| | - Moshi Song
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China.
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, 100101, China.
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, 100101, China.
- University of Chinese Academy of Sciences, Beijing, 100049, China.
| | - Jing-Dong J Han
- Peking-Tsinghua Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, Center for Quantitative Biology, Peking University, Beijing, 100871, China.
| | - Guang-Hui Liu
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China.
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, 100101, China.
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, 100101, China.
- University of Chinese Academy of Sciences, Beijing, 100049, China.
- Advanced Innovation Center for Human Brain Protection, National Clinical Research Center for Geriatric Disorders, Xuanwu Hospital Capital Medical University, Beijing, 100053, China.
- Aging Translational Medicine Center, International Center for Aging and Cancer, Beijing Municipal Geriatric Medical Research Center, Xuanwu Hospital Capital Medical University, Beijing, 100053, China.
| |
Collapse
|
14
|
Shosha E, Qin L, Lemtalsi T, Zaidi SAH, Rojas M, Xu Z, Caldwell RW, Caldwell RB, Fouda AY. Investigation of Retinal Metabolic Function in Type 1 Diabetic Akita Mice. Front Cardiovasc Med 2022; 9:900640. [PMID: 35722112 PMCID: PMC9201036 DOI: 10.3389/fcvm.2022.900640] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Accepted: 05/16/2022] [Indexed: 12/14/2022] Open
Abstract
Diabetic retinopathy (DR) is the leading cause of vision loss in working age adults. Understanding the retinal metabolic response to circulating high glucose levels in diabetic patients is critical for development of new therapeutics to treat DR. Measuring retinal metabolic function using the Seahorse analyzer is a promising technique to investigate the effect of hyperglycemia on retinal glycolysis and mitochondrial respiration. Here, we analyzed the retinal metabolic function in young and old diabetic and control mice. We also compared the expression of key glycolytic enzymes between the two groups. The Seahorse XF analyzer was used to measure the metabolic function of retina explants from young and old type 1 diabetic Akita (Ins2Akita) mice and their control littermates. Rate-limiting glycolytic enzymes were analyzed in retina lysates from the two age groups by Western blotting. Retinas from young adult Akita mice showed a decreased glycolytic response as compared to control littermates. However, this was not observed in the older mice. Western blotting analysis showed decreased expression of the glycolytic enzyme PFKFB3 in the young Akita mice retinas. Measurement of the oxygen consumption rate showed no difference in retinal mitochondrial respiration between Akita and WT littermates under normal glucose conditions ex vivo despite mitochondrial fragmentation in the Akita retinas as examined by electron microscopy. However, Akita mice retinas showed decreased mitochondrial respiration under glucose-free conditions. In conclusion, diabetic retinas display a decreased glycolytic response during the early course of diabetes which is accompanied by a reduction in PFKFB3. Diabetic retinas exhibit decreased mitochondrial respiration under glucose deprivation.
Collapse
Affiliation(s)
- Esraa Shosha
- Vascular Biology Center, Augusta University, Augusta, GA, United States
- Department of Clinical Pharmacy, Faculty of Pharmacy, Cairo University, Giza, Egypt
- University of Arkansas for Medical Sciences, Little Rock, AR, United States
| | - Luke Qin
- Vascular Biology Center, Augusta University, Augusta, GA, United States
| | - Tahira Lemtalsi
- Vascular Biology Center, Augusta University, Augusta, GA, United States
- Culver Vision Discovery Institute, Augusta University, Augusta, GA, United States
| | - Syed A. H. Zaidi
- Vascular Biology Center, Augusta University, Augusta, GA, United States
- Culver Vision Discovery Institute, Augusta University, Augusta, GA, United States
| | - Modesto Rojas
- Vascular Biology Center, Augusta University, Augusta, GA, United States
- Culver Vision Discovery Institute, Augusta University, Augusta, GA, United States
| | - Zhimin Xu
- Vascular Biology Center, Augusta University, Augusta, GA, United States
- Culver Vision Discovery Institute, Augusta University, Augusta, GA, United States
| | - Robert William Caldwell
- Culver Vision Discovery Institute, Augusta University, Augusta, GA, United States
- Department of Pharmacology and Toxicology, Augusta University, Augusta, GA, United States
| | - Ruth B. Caldwell
- Vascular Biology Center, Augusta University, Augusta, GA, United States
- Culver Vision Discovery Institute, Augusta University, Augusta, GA, United States
- *Correspondence: Ruth B. Caldwell,
| | - Abdelrahman Y. Fouda
- Department of Clinical Pharmacy, Faculty of Pharmacy, Cairo University, Giza, Egypt
- University of Arkansas for Medical Sciences, Little Rock, AR, United States
- Abdelrahman Y. Fouda,
| |
Collapse
|
15
|
McLaughlin T, Medina A, Perkins J, Yera M, Wang JJ, Zhang SX. Cellular stress signaling and the unfolded protein response in retinal degeneration: mechanisms and therapeutic implications. Mol Neurodegener 2022; 17:25. [PMID: 35346303 PMCID: PMC8962104 DOI: 10.1186/s13024-022-00528-w] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Accepted: 03/04/2022] [Indexed: 12/22/2022] Open
Abstract
Background The retina, as part of the central nervous system (CNS) with limited capacity for self-reparation and regeneration in mammals, is under cumulative environmental stress due to high-energy demands and rapid protein turnover. These stressors disrupt the cellular protein and metabolic homeostasis, which, if not alleviated, can lead to dysfunction and cell death of retinal neurons. One primary cellular stress response is the highly conserved unfolded protein response (UPR). The UPR acts through three main signaling pathways in an attempt to restore the protein homeostasis in the endoplasmic reticulum (ER) by various means, including but not limited to, reducing protein translation, increasing protein-folding capacity, and promoting misfolded protein degradation. Moreover, recent work has identified a novel function of the UPR in regulation of cellular metabolism and mitochondrial function, disturbance of which contributes to neuronal degeneration and dysfunction. The role of the UPR in retinal neurons during aging and under disease conditions in age-related macular degeneration (AMD), retinitis pigmentosa (RP), glaucoma, and diabetic retinopathy (DR) has been explored over the past two decades. Each of the disease conditions and their corresponding animal models provide distinct challenges and unique opportunities to gain a better understanding of the role of the UPR in the maintenance of retinal health and function. Method We performed an extensive literature search on PubMed and Google Scholar using the following keywords: unfolded protein response, metabolism, ER stress, retinal degeneration, aging, age-related macular degeneration, retinitis pigmentosa, glaucoma, diabetic retinopathy. Results and conclusion We summarize recent advances in understanding cellular stress response, in particular the UPR, in retinal diseases, highlighting the potential roles of UPR pathways in regulation of cellular metabolism and mitochondrial function in retinal neurons. Further, we provide perspective on the promise and challenges for targeting the UPR pathways as a new therapeutic approach in age- and disease-related retinal degeneration.
Collapse
Affiliation(s)
- Todd McLaughlin
- Department of Ophthalmology and Ira G. Ross Eye Institute, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, State University of New York, 955 Main Street, Buffalo, NY, 14203, USA
| | - Andy Medina
- Department of Ophthalmology and Ira G. Ross Eye Institute, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, State University of New York, 955 Main Street, Buffalo, NY, 14203, USA
| | - Jacob Perkins
- Department of Ophthalmology and Ira G. Ross Eye Institute, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, State University of New York, 955 Main Street, Buffalo, NY, 14203, USA
| | - Maria Yera
- Department of Ophthalmology and Ira G. Ross Eye Institute, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, State University of New York, 955 Main Street, Buffalo, NY, 14203, USA.,Neuroscience Program, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, State University of New York, Buffalo, NY, USA
| | - Joshua J Wang
- Department of Ophthalmology and Ira G. Ross Eye Institute, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, State University of New York, 955 Main Street, Buffalo, NY, 14203, USA.,Neuroscience Program, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, State University of New York, Buffalo, NY, USA
| | - Sarah X Zhang
- Department of Ophthalmology and Ira G. Ross Eye Institute, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, State University of New York, 955 Main Street, Buffalo, NY, 14203, USA. .,Neuroscience Program, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, State University of New York, Buffalo, NY, USA. .,Department of Biochemistry, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, State University of New York, Buffalo, NY, USA.
| |
Collapse
|
16
|
Conte M, Giuliani C, Chiariello A, Iannuzzi V, Franceschi C, Salvioli S. GDF15, an emerging key player in human aging. Ageing Res Rev 2022; 75:101569. [PMID: 35051643 DOI: 10.1016/j.arr.2022.101569] [Citation(s) in RCA: 57] [Impact Index Per Article: 28.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Accepted: 01/14/2022] [Indexed: 12/20/2022]
Abstract
Growth differentiation factor 15 (GDF15) is recently emerging not only as a stress-related mitokine, but also as a key player in the aging process, being one of the most up-regulated protein with age and associated with a variety of age-related diseases (ARDs). Many data indicate that GDF15 has protective roles in several tissues during different stress and aging, thus playing a beneficial role in apparent contrast with the observed association with many ARDs. A possible detrimental role for this protein is then hypothesized to emerge with age. Therefore, GDF15 can be considered as a pleiotropic factor with beneficial activities that can turn detrimental in old age possibly when it is chronically elevated. In this review, we summarize the current knowledge on the biology of GDF15 during aging. We also propose GDF15 as a part of a dormancy program, where it may play a role as a mediator of defense processes aimed to protect from inflammatory damage and other stresses, according to the life history theory.
Collapse
Affiliation(s)
- Maria Conte
- Department of Experimental, Diagnostic and Specialty Medicine (DIMES), University of Bologna, Bologna, Italy; Interdepartmental Centre "Alma Mater Research Institute on Global Challenges and Climate Change (Alma Climate)", University of Bologna, Bologna, Italy.
| | - Cristina Giuliani
- Interdepartmental Centre "Alma Mater Research Institute on Global Challenges and Climate Change (Alma Climate)", University of Bologna, Bologna, Italy; Laboratory of Molecular Anthropology & Centre for Genome Biology, Department of Biological, Geological and Environmental Sciences, University of Bologna, Bologna, Italy
| | - Antonio Chiariello
- Department of Experimental, Diagnostic and Specialty Medicine (DIMES), University of Bologna, Bologna, Italy
| | - Vincenzo Iannuzzi
- Laboratory of Molecular Anthropology & Centre for Genome Biology, Department of Biological, Geological and Environmental Sciences, University of Bologna, Bologna, Italy
| | - Claudio Franceschi
- Department of Experimental, Diagnostic and Specialty Medicine (DIMES), University of Bologna, Bologna, Italy; Institute of Information Technologies, Mathematics and Mechanics, Lobachevsky University, Nizhniy Novgorod, Russia
| | - Stefano Salvioli
- Department of Experimental, Diagnostic and Specialty Medicine (DIMES), University of Bologna, Bologna, Italy; Interdepartmental Centre "Alma Mater Research Institute on Global Challenges and Climate Change (Alma Climate)", University of Bologna, Bologna, Italy
| |
Collapse
|
17
|
Li R, Li Y, Tian M, Zhang H, Lou L, Liu K, Zhang J, Zhao Y, Zhang J, Le S, Fu X, Zhou Y, Li W, Gao X, Nie Y. Comparative proteomic profiling reveals a pathogenic role for the O-GlcNAcylated AIMP2-PARP1 complex in aging-related hepatic steatosis in mice. FEBS Lett 2022; 596:128-145. [PMID: 34817071 DOI: 10.1002/1873-3468.14242] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2021] [Revised: 10/29/2021] [Accepted: 11/15/2021] [Indexed: 11/07/2022]
Abstract
The prevalence of non-alcoholic fatty liver disease (NAFLD) increases with aging. However, the mechanism of aging-related NAFLD remains unclear. Herein, we constructed an aging-related hepatic steatosis model and analyzed the differentially expressed proteins (DEPs) in livers from young and old mice using liquid chromatography-mass spectrometry. Five hundred and eighty-eight aging-related DEPs and novel pathways were identified. Aminoacyl tRNA synthetase complex-interacting multifunctional protein 2 (AIMP2), the most significantly upregulated protein, promoted poly(ADP-ribose) polymerase 1 (PARP1) activation in aging-related hepatic steatosis. Additionally, mice liver-specific O-GlcNAcase knockout promoted AIMP2 and PARP1 expression. O-GlcNAc transferase (OGT) overexpression and O-GlcNAcase inhibition by genetic or pharmaceutical manipulations increased AIMP2 and PARP1 levels in vitro. Mechanistically, O-GlcNAcylation increased AIMP2 protein stability, leading to its aggregation. Our study reveals O-GlcNAcylated AIMP2 as a novel pathogenic regulator of aging-related hepatic steatosis.
Collapse
Affiliation(s)
- Renlong Li
- State key Laboratory of Cancer Biology, National Clinical Research Center for Digestive Diseases and Xijing Hospital of Digestive Diseases, Air Force Military Medical University, Xi'an, China
| | - Yan Li
- State key Laboratory of Cancer Biology, National Clinical Research Center for Digestive Diseases and Xijing Hospital of Digestive Diseases, Air Force Military Medical University, Xi'an, China
| | - Miaomiao Tian
- State key Laboratory of Cancer Biology, National Clinical Research Center for Digestive Diseases and Xijing Hospital of Digestive Diseases, Air Force Military Medical University, Xi'an, China
| | - Haohao Zhang
- State key Laboratory of Cancer Biology, National Clinical Research Center for Digestive Diseases and Xijing Hospital of Digestive Diseases, Air Force Military Medical University, Xi'an, China
| | - Lijun Lou
- State key Laboratory of Cancer Biology, National Clinical Research Center for Digestive Diseases and Xijing Hospital of Digestive Diseases, Air Force Military Medical University, Xi'an, China
| | - Kun Liu
- State key Laboratory of Cancer Biology, National Clinical Research Center for Digestive Diseases and Xijing Hospital of Digestive Diseases, Air Force Military Medical University, Xi'an, China
| | - Jiehao Zhang
- State key Laboratory of Cancer Biology, National Clinical Research Center for Digestive Diseases and Xijing Hospital of Digestive Diseases, Air Force Military Medical University, Xi'an, China
| | - Yu Zhao
- State key Laboratory of Cancer Biology, National Clinical Research Center for Digestive Diseases and Xijing Hospital of Digestive Diseases, Air Force Military Medical University, Xi'an, China
| | - Jing Zhang
- State key Laboratory of Cancer Biology, National Clinical Research Center for Digestive Diseases and Xijing Hospital of Digestive Diseases, Air Force Military Medical University, Xi'an, China
| | - Shuangshuang Le
- State key Laboratory of Cancer Biology, National Clinical Research Center for Digestive Diseases and Xijing Hospital of Digestive Diseases, Air Force Military Medical University, Xi'an, China
| | - Xin Fu
- State key Laboratory of Cancer Biology, National Clinical Research Center for Digestive Diseases and Xijing Hospital of Digestive Diseases, Air Force Military Medical University, Xi'an, China
| | - Yao Zhou
- State key Laboratory of Cancer Biology, National Clinical Research Center for Digestive Diseases and Xijing Hospital of Digestive Diseases, Air Force Military Medical University, Xi'an, China
| | - Wenjiao Li
- State key Laboratory of Cancer Biology, National Clinical Research Center for Digestive Diseases and Xijing Hospital of Digestive Diseases, Air Force Military Medical University, Xi'an, China
| | - Xianchun Gao
- State key Laboratory of Cancer Biology, National Clinical Research Center for Digestive Diseases and Xijing Hospital of Digestive Diseases, Air Force Military Medical University, Xi'an, China
| | - Yongzhan Nie
- State key Laboratory of Cancer Biology, National Clinical Research Center for Digestive Diseases and Xijing Hospital of Digestive Diseases, Air Force Military Medical University, Xi'an, China
| |
Collapse
|
18
|
Zmyslowska A, Kuljanin M, Malachowska B, Stanczak M, Michalek D, Wlodarczyk A, Grot D, Taha J, Pawlik B, Lebiedzińska-Arciszewska M, Nieznanska H, Wieckowski MR, Rieske P, Mancias JD, Borowiec M, Mlynarski W, Fendler W. Multiomic analysis on human cell model of wolfram syndrome reveals changes in mitochondrial morphology and function. Cell Commun Signal 2021; 19:116. [PMID: 34801048 PMCID: PMC8605533 DOI: 10.1186/s12964-021-00791-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Accepted: 10/01/2021] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND Wolfram syndrome (WFS) is a rare autosomal recessive syndrome in which diabetes mellitus and neurodegenerative disorders occur as a result of Wolframin deficiency and increased ER stress. In addition, WFS1 deficiency leads to calcium homeostasis disturbances and can change mitochondrial dynamics. The aim of this study was to evaluate protein levels and changes in gene transcription on human WFS cell model under experimental ER stress. METHODS We performed transcriptomic and proteomic analysis on WFS human cell model-skin fibroblasts reprogrammed into induced pluripotent stem (iPS) cells and then into neural stem cells (NSC) with subsequent ER stress induction using tunicamycin (TM). Results were cross-referenced with publicly available RNA sequencing data in hippocampi and hypothalami of mice with WFS1 deficiency. RESULTS Proteomic analysis identified specific signal pathways that differ in NSC WFS cells from healthy ones. Next, detailed analysis of the proteins involved in the mitochondrial function showed the down-regulation of subunits of the respiratory chain complexes in NSC WFS cells, as well as the up-regulation of proteins involved in Krebs cycle and glycolysis when compared to the control cells. Based on pathway enrichment analysis we concluded that in samples from mice hippocampi the mitochondrial protein import machinery and OXPHOS were significantly down-regulated. CONCLUSIONS Our results show the functional and morphological secondary mitochondrial damage in patients with WFS. Video Abstract.
Collapse
Affiliation(s)
- Agnieszka Zmyslowska
- Department of Clinical Genetics, Medical University of Lodz, Pomorska Str. 251, 92-213 Lodz, Poland
| | - Miljan Kuljanin
- Department of Radiation Oncology, Dana-Farber Cancer Institute, Boston, MA USA
| | - Beata Malachowska
- Department of Biostatistics and Translational Medicine, Medical University of Lodz, Lodz, Poland
- Department of Radiation Oncology, Einstein College of Medicine, Bronx, NY USA
| | - Marcin Stanczak
- Department of Biostatistics and Translational Medicine, Medical University of Lodz, Lodz, Poland
| | - Dominika Michalek
- Department of Biostatistics and Translational Medicine, Medical University of Lodz, Lodz, Poland
| | - Aneta Wlodarczyk
- Department of Tumor Biology, Medical University of Lodz, Lodz, Poland
| | - Dagmara Grot
- Department of Tumor Biology, Medical University of Lodz, Lodz, Poland
| | - Joanna Taha
- Central Laboratory for Genetic Research in Pediatric Oncology “Oncolab”, Medical University of Lodz, Lodz, Poland
| | - Bartłomiej Pawlik
- Department of Pediatrics, Oncology and Hematology, Medical University of Lodz, Lodz, Poland
- Postgraduate School of Molecular Medicine, Medical University of Warsaw, Warsaw, Poland
| | | | - Hanna Nieznanska
- Nencki Institute of Experimental Biology, Polish Academy of Sciences, Warsaw, Poland
| | - Mariusz R. Wieckowski
- Nencki Institute of Experimental Biology, Polish Academy of Sciences, Warsaw, Poland
| | - Piotr Rieske
- Department of Tumor Biology, Medical University of Lodz, Lodz, Poland
| | - Joseph D. Mancias
- Department of Radiation Oncology, Dana-Farber Cancer Institute, Boston, MA USA
| | - Maciej Borowiec
- Department of Clinical Genetics, Medical University of Lodz, Pomorska Str. 251, 92-213 Lodz, Poland
| | - Wojciech Mlynarski
- Department of Pediatrics, Oncology and Hematology, Medical University of Lodz, Lodz, Poland
| | - Wojciech Fendler
- Department of Radiation Oncology, Dana-Farber Cancer Institute, Boston, MA USA
- Department of Biostatistics and Translational Medicine, Medical University of Lodz, Lodz, Poland
| |
Collapse
|
19
|
Yang TT, Li H, Dong LJ. Role of glycolysis in retinal vascular endothelium, glia, pigment epithelium, and photoreceptor cells and as therapeutic targets for related retinal diseases. Int J Ophthalmol 2021; 14:1302-1309. [PMID: 34540603 DOI: 10.18240/ijo.2021.09.02] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Accepted: 06/16/2021] [Indexed: 02/08/2023] Open
Abstract
Glycolysis produces large amounts of adenosine triphosphate (ATP) in a short time. The retinal vascular endothelium feeds itself primarily through aerobic glycolysis with less ATP. But when it generates new vessels, aerobic glycolysis provides rapid and abundant ATP support for angiogenesis, and thus inhibition of glycolysis in endothelial cells can be a target for the treatment of neovascularization. Aerobic glycolysis has a protective effect on Müller cells, and it can provide with a target for visual protection and maintenance of the blood-retinal barrier. Under physiological conditions, the mitochondria of RPE can use lactic acid produced by photoreceptor cells as an energy source to provide ATP for survival. In pathological conditions, because RPE cells avoid their oxidative damage by increasing glycolysis, a large number of glycolysis products accumulate, which in turn has a toxic effect on photoreceptor cells. This shows that stabilizing the function of RPE mitochondria may become a target for the treatment of diseases such as retinal degeneration. The decrease of aerobic glycolysis leads to the decline of photoreceptor cell function and impaired vision; therefore, aerobic glycolysis of stable photoreceptor cells provides a reliable target for delaying vision loss. It is of great significance to study the role of glycolysis in various retinal cells for the targeted treatment of ocular fundus diseases.
Collapse
Affiliation(s)
- Ting-Ting Yang
- Editorial Department of Chinese Journal of Ocular Fundus Diseases, West China Hospital of Sichuan University, Chengdu 610041, Sichuan Province, China
| | - Hui Li
- Tianjin Key Laboratory of Retinal Functions and Diseases, Tianjin Branch of National Clinical Research Center for Ocular Diseases, Eye Institute and School of Optometry, Tianjin Medical University Eye Hospital, Tianjin 300384, China
| | - Li-Jie Dong
- Tianjin Key Laboratory of Retinal Functions and Diseases, Tianjin Branch of National Clinical Research Center for Ocular Diseases, Eye Institute and School of Optometry, Tianjin Medical University Eye Hospital, Tianjin 300384, China
| |
Collapse
|
20
|
Campello L, Singh N, Advani J, Mondal AK, Corso-Díaz X, Swaroop A. Aging of the Retina: Molecular and Metabolic Turbulences and Potential Interventions. Annu Rev Vis Sci 2021; 7:633-664. [PMID: 34061570 PMCID: PMC11375453 DOI: 10.1146/annurev-vision-100419-114940] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Multifaceted and divergent manifestations across tissues and cell types have curtailed advances in deciphering the cellular events that accompany advanced age and contribute to morbidities and mortalities. Increase in human lifespan during the past century has heightened awareness of the need to prevent age-associated frailty of neuronal and sensory systems to allow a healthy and productive life. In this review, we discuss molecular and physiological attributes of aging of the retina, with a goal of understanding age-related impairment of visual function. We highlight the epigenome-metabolism nexus and proteostasis as key contributors to retinal aging and discuss lifestyle changes as potential modulators of retinal function. Finally, we deliberate promising intervention strategies for promoting healthy aging of the retina for improved vision.
Collapse
Affiliation(s)
- Laura Campello
- Neurobiology, Neurodegeneration and Repair Laboratory, National Eye Institute, National Institutes of Health, Bethesda, Maryland 20892, USA;
| | - Nivedita Singh
- Neurobiology, Neurodegeneration and Repair Laboratory, National Eye Institute, National Institutes of Health, Bethesda, Maryland 20892, USA;
| | - Jayshree Advani
- Neurobiology, Neurodegeneration and Repair Laboratory, National Eye Institute, National Institutes of Health, Bethesda, Maryland 20892, USA;
| | - Anupam K Mondal
- Neurobiology, Neurodegeneration and Repair Laboratory, National Eye Institute, National Institutes of Health, Bethesda, Maryland 20892, USA;
| | - Ximena Corso-Díaz
- Neurobiology, Neurodegeneration and Repair Laboratory, National Eye Institute, National Institutes of Health, Bethesda, Maryland 20892, USA;
| | - Anand Swaroop
- Neurobiology, Neurodegeneration and Repair Laboratory, National Eye Institute, National Institutes of Health, Bethesda, Maryland 20892, USA;
| |
Collapse
|
21
|
Bilbao-Malavé V, González-Zamora J, de la Puente M, Recalde S, Fernandez-Robredo P, Hernandez M, Layana AG, Saenz de Viteri M. Mitochondrial Dysfunction and Endoplasmic Reticulum Stress in Age Related Macular Degeneration, Role in Pathophysiology, and Possible New Therapeutic Strategies. Antioxidants (Basel) 2021; 10:1170. [PMID: 34439418 PMCID: PMC8388889 DOI: 10.3390/antiox10081170] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Revised: 07/14/2021] [Accepted: 07/20/2021] [Indexed: 02/07/2023] Open
Abstract
Age related macular degeneration (AMD) is the main cause of legal blindness in developed countries. It is a multifactorial disease in which a combination of genetic and environmental factors contributes to increased risk of developing this vision-incapacitating condition. Oxidative stress plays a central role in the pathophysiology of AMD and recent publications have highlighted the importance of mitochondrial dysfunction and endoplasmic reticulum stress in this disease. Although treatment with vascular endothelium growth factor inhibitors have decreased the risk of blindness in patients with the exudative form of AMD, the search for new therapeutic options continues to prevent the loss of photoreceptors and retinal pigment epithelium cells, characteristic of late stage AMD. In this review, we explain how mitochondrial dysfunction and endoplasmic reticulum stress participate in AMD pathogenesis. We also discuss a role of several antioxidants (bile acids, resveratrol, melatonin, humanin, and coenzyme Q10) in amelioration of AMD pathology.
Collapse
Affiliation(s)
- Valentina Bilbao-Malavé
- Department of Opthalmology, Clínica Universidad de Navarra, 31008 Pamplona, Spain; (V.B.-M.); (J.G.-Z.); (M.d.l.P.); (A.G.L.)
| | - Jorge González-Zamora
- Department of Opthalmology, Clínica Universidad de Navarra, 31008 Pamplona, Spain; (V.B.-M.); (J.G.-Z.); (M.d.l.P.); (A.G.L.)
| | - Miriam de la Puente
- Department of Opthalmology, Clínica Universidad de Navarra, 31008 Pamplona, Spain; (V.B.-M.); (J.G.-Z.); (M.d.l.P.); (A.G.L.)
| | - Sergio Recalde
- Retinal Pathologies and New Therapies Group, Experimental Ophthalmology Laboratory, Department of Ophthalmology, Universidad de Navarra, 31008 Pamplona, Spain; (S.R.); (P.F.-R.); (M.H.)
- Navarra Institute for Health Research, IdiSNA, 31008 Pamplona, Spain
- Red Temática de Investigación Cooperativa en Salud: ‘Prevention, Early Detection, and Treatment of the Prevalent Degenerative and Chronic Ocular Pathology’ from (RD16/0008/0011), Ministerio de Ciencia, Innovación y Universidades, Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Patricia Fernandez-Robredo
- Retinal Pathologies and New Therapies Group, Experimental Ophthalmology Laboratory, Department of Ophthalmology, Universidad de Navarra, 31008 Pamplona, Spain; (S.R.); (P.F.-R.); (M.H.)
- Navarra Institute for Health Research, IdiSNA, 31008 Pamplona, Spain
- Red Temática de Investigación Cooperativa en Salud: ‘Prevention, Early Detection, and Treatment of the Prevalent Degenerative and Chronic Ocular Pathology’ from (RD16/0008/0011), Ministerio de Ciencia, Innovación y Universidades, Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - María Hernandez
- Retinal Pathologies and New Therapies Group, Experimental Ophthalmology Laboratory, Department of Ophthalmology, Universidad de Navarra, 31008 Pamplona, Spain; (S.R.); (P.F.-R.); (M.H.)
- Navarra Institute for Health Research, IdiSNA, 31008 Pamplona, Spain
- Red Temática de Investigación Cooperativa en Salud: ‘Prevention, Early Detection, and Treatment of the Prevalent Degenerative and Chronic Ocular Pathology’ from (RD16/0008/0011), Ministerio de Ciencia, Innovación y Universidades, Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Alfredo Garcia Layana
- Department of Opthalmology, Clínica Universidad de Navarra, 31008 Pamplona, Spain; (V.B.-M.); (J.G.-Z.); (M.d.l.P.); (A.G.L.)
- Retinal Pathologies and New Therapies Group, Experimental Ophthalmology Laboratory, Department of Ophthalmology, Universidad de Navarra, 31008 Pamplona, Spain; (S.R.); (P.F.-R.); (M.H.)
- Navarra Institute for Health Research, IdiSNA, 31008 Pamplona, Spain
- Red Temática de Investigación Cooperativa en Salud: ‘Prevention, Early Detection, and Treatment of the Prevalent Degenerative and Chronic Ocular Pathology’ from (RD16/0008/0011), Ministerio de Ciencia, Innovación y Universidades, Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Manuel Saenz de Viteri
- Department of Opthalmology, Clínica Universidad de Navarra, 31008 Pamplona, Spain; (V.B.-M.); (J.G.-Z.); (M.d.l.P.); (A.G.L.)
- Retinal Pathologies and New Therapies Group, Experimental Ophthalmology Laboratory, Department of Ophthalmology, Universidad de Navarra, 31008 Pamplona, Spain; (S.R.); (P.F.-R.); (M.H.)
- Navarra Institute for Health Research, IdiSNA, 31008 Pamplona, Spain
- Red Temática de Investigación Cooperativa en Salud: ‘Prevention, Early Detection, and Treatment of the Prevalent Degenerative and Chronic Ocular Pathology’ from (RD16/0008/0011), Ministerio de Ciencia, Innovación y Universidades, Instituto de Salud Carlos III, 28029 Madrid, Spain
| |
Collapse
|
22
|
Soldatov VO, Kukharsky MS, Belykh AE, Sobolev AM, Deykin AV. Retinal Damage in Amyotrophic Lateral Sclerosis: Underlying Mechanisms. Eye Brain 2021; 13:131-146. [PMID: 34012311 PMCID: PMC8128130 DOI: 10.2147/eb.s299423] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2021] [Accepted: 03/04/2021] [Indexed: 01/04/2023] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a fatal neurodegenerative disease resulting in a gradual loss of motor neuron function. Although ophthalmic complaints are not presently considered a classic symptom of ALS, retinal changes such as thinning, axonal degeneration and inclusion bodies have been found in many patients. Retinal abnormalities observed in postmortem human tissues and animal models are similar to spinal cord changes in ALS. These findings are not dramatically unexpected because retina shares an ontogenetic relationship with the brain, and many genes are associated both with neurodegeneration and retinal diseases. Experimental studies have demonstrated that ALS affects many “vulnerable points” of the retina. Aggregate deposition, impaired nuclear protein import, endoplasmic reticulum stress, glutamate excitotoxicity, vascular regression, and mitochondrial dysfunction are factors suspected as being the main cause of motor neuron damage in ALS. Herein, we show that all of these pathways can affect retinal cells in the same way as motor neurons. Furthermore, we suppose that understanding the patterns of neuro-ophthalmic interaction in ALS can help in the diagnosis and treatment of this disease.
Collapse
Affiliation(s)
- Vladislav O Soldatov
- Core Facility Centre, Institute of Gene Biology, Russian Academy of Sciences, Moscow, Russia.,Department of Pharmacology and Clinical Pharmacology, Belgorod State National Research University, Belgorod, Russia
| | - Michail S Kukharsky
- Department of General and Cell Biology, Faculty of Medical Biology, Pirogov Russian National Research Medical University, Moscow, Russia.,Laboratory of Genetic Modelling of Neurodegenerative Processes, Institute of Physiologically Active Compounds, Russian Academy of Sciences, Chernogolovka, Russia
| | - Andrey E Belykh
- Department of Pathophysiology, Kursk State Medical University, Kursk, Russia
| | - Andrey M Sobolev
- Laboratory of Genetic Modelling of Neurodegenerative Processes, Institute of Physiologically Active Compounds, Russian Academy of Sciences, Chernogolovka, Russia
| | - Alexey V Deykin
- Department of Pharmacology and Clinical Pharmacology, Belgorod State National Research University, Belgorod, Russia.,Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Institute of Gene Biology, Russian Academy of Sciences, Moscow, Russia
| |
Collapse
|
23
|
Li H, Liu B, Lian L, Zhou J, Xiang S, Zhai Y, Chen Y, Ma X, Wu W, Hou L. High dose expression of heme oxigenase-1 induces retinal degeneration through ER stress-related DDIT3. Mol Neurodegener 2021; 16:16. [PMID: 33691741 PMCID: PMC7944639 DOI: 10.1186/s13024-021-00437-4] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Accepted: 03/01/2021] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Oxidative stress is a common cause of neurodegeneration and plays a central role in retinal degenerative diseases. Heme oxygenase-1 (HMOX1) is a redox-regulated enzyme that is induced in neurodegenerative diseases and acts against oxidative stress but can also promote cell death, a phenomenon that is still unexplained in molecular terms. Here, we test whether HMOX1 has opposing effects during retinal degeneration and investigate the molecular mechanisms behind its pro-apoptotic role. METHODS Basal and induced levels of HMOX1 in retinas are examined during light-induced retinal degeneration in mice. Light damage-independent HMOX1 induction at two different expression levels is achieved by intraocular injection of different doses of an adeno-associated virus vector expressing HMOX1. Activation of Müller glial cells, retinal morphology and photoreceptor cell death are examined using hematoxylin-eosin staining, TUNEL assays, immunostaining and retinal function are evaluated with electroretinograms. Downstream gene expression of HMOX1 is analyzed by RNA-seq, qPCR examination and western blotting. The role of one of these genes, the pro-apoptotic DNA damage inducible transcript 3 (Ddit3), is analyzed in a line of knockout mice. RESULTS Light-induced retinal degeneration leads to photoreceptor degeneration and concomitant HMOX1 induction. HMOX1 expression at low levels before light exposure prevents photoreceptor degeneration but expression at high levels directly induces photoreceptor degeneration even without light stress. Photoreceptor degeneration following high level expression of HMOX1 is associated with a mislocalization of rhodopsin in photoreceptors and an increase in the expression of DDIT3. Genetic deletion of Ddit3 in knockout mice prevents photoreceptor cell degeneration normally resulting from high level HMOX1 expression. CONCLUSION The results reveal that the expression levels determine whether HMOX1 is protective or deleterious in the retina. Furthermore, in contrast to the protective low dose of HMOX1, the deleterious high dose is associated with induction of DDIT3 and endoplasmic reticulum stress as manifested, for instance, in rhodopsin mislocalization. Hence, future applications of HMOX1 or its regulated targets in gene therapy approaches should carefully consider expression levels in order to avoid potentially devastating effects.
Collapse
Affiliation(s)
- Huirong Li
- Laboratory of Developmental Cell Biology and Disease, School of Ophthalmology and Optometry and Eye Hospital, and State Key Laboratory of Ophthalmology, Optometry and Vision Science, Wenzhou Medical University, Wenzhou, China
| | - Bo Liu
- Laboratory of Developmental Cell Biology and Disease, School of Ophthalmology and Optometry and Eye Hospital, and State Key Laboratory of Ophthalmology, Optometry and Vision Science, Wenzhou Medical University, Wenzhou, China
| | - Lili Lian
- Laboratory of Developmental Cell Biology and Disease, School of Ophthalmology and Optometry and Eye Hospital, and State Key Laboratory of Ophthalmology, Optometry and Vision Science, Wenzhou Medical University, Wenzhou, China
| | - Jiajia Zhou
- Laboratory of Developmental Cell Biology and Disease, School of Ophthalmology and Optometry and Eye Hospital, and State Key Laboratory of Ophthalmology, Optometry and Vision Science, Wenzhou Medical University, Wenzhou, China
| | - Shengjin Xiang
- Eye Hospital of Wenzhou Medical University, Wenzhou, 325003 China
| | - Yifan Zhai
- Laboratory of Developmental Cell Biology and Disease, School of Ophthalmology and Optometry and Eye Hospital, and State Key Laboratory of Ophthalmology, Optometry and Vision Science, Wenzhou Medical University, Wenzhou, China
| | - Yu Chen
- Laboratory of Developmental Cell Biology and Disease, School of Ophthalmology and Optometry and Eye Hospital, and State Key Laboratory of Ophthalmology, Optometry and Vision Science, Wenzhou Medical University, Wenzhou, China
| | - Xiaoyin Ma
- Laboratory of Developmental Cell Biology and Disease, School of Ophthalmology and Optometry and Eye Hospital, and State Key Laboratory of Ophthalmology, Optometry and Vision Science, Wenzhou Medical University, Wenzhou, China
| | - Wencan Wu
- Eye Hospital of Wenzhou Medical University, Wenzhou, 325003 China
| | - Ling Hou
- Laboratory of Developmental Cell Biology and Disease, School of Ophthalmology and Optometry and Eye Hospital, and State Key Laboratory of Ophthalmology, Optometry and Vision Science, Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
24
|
Long P, He M, Yan W, Chen W, Wei D, Wang S, Zhang Z, Ge W, Chen T. ALDH2 protects naturally aged mouse retina via inhibiting oxidative stress-related apoptosis and enhancing unfolded protein response in endoplasmic reticulum. Aging (Albany NY) 2020; 13:2750-2767. [PMID: 33411685 PMCID: PMC7880320 DOI: 10.18632/aging.202325] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2020] [Accepted: 10/01/2020] [Indexed: 12/14/2022]
Abstract
During the process of aging, the retina exhibits chronic oxidative stress (OS) damage. Our preliminary experiment showed that acetaldehyde dehydrogenase 2 (ALDH2) could alleviate retinal damage caused by OS. This study aimed to explore whether ALDH2 could inhibit mice retinal cell apoptosis and enhance the function of unfolded protein response in endoplasmic reticulum (UPRER) through reducing OS in aging process. Retinal function and structure in vivo and in vitro were examined in aged ALDH2+ overexpression mice and ALDH2 agonist Alda1-treated aged mice. Levels of ALDH2, endoplasmic reticulum stress (ERS), apoptosis and inflammatory cytokines were evaluated. Higher expression of ALDH2 was observed at the outer nuclear layer (ONL) and the inner nuclear layer (INL) in aged ALDH2+ overexpression and aged Alda1-treated mice. Moreover, aged ALDH2+ overexpression mice and aged Alda1-treated mice exhibited better retinal function and structure. Increased expression of glucose-regulated protein 78 (GRP78) and ERS-related protein phosphorylated eukaryotic initiation factor 2 (peIF2α) and decreased expression of apoptosis-related protein, including C/EBP homologous protein (CHOP), caspase12 and caspase9, and retinal inflammatory cytokines were detected in the retina of aged ALDH2+ overexpression mice and aged Alda1-treated mice. The expression of ALDH2 in the retina was decreased in aging process. ALDH2 could reduce retinal oxidative stress and apoptosis, strengthen UPRER during the aging process to improve retinal function and structure.
Collapse
Affiliation(s)
- Pan Long
- Department of General Practice, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, Shaanxi Province, China.,Department of Ophthalmology, The General Hospital of Western Theater Command, Chengdu 610083, Sichuan Province, China.,Center of Clinical Aerospace Medicine, Fourth Military Medical University, Xi'an 710032, Shaanxi Province, China
| | - Mengshan He
- Department of Chinese Material Medical and Natural Medicines, Fourth Military Medical University, Xi'an 710032, Shaanxi Province, China.,Department of Pharmacy, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, Shaanxi Province, China
| | - Weiming Yan
- Department of Ophthalmology, The 900th Hospital of Joint Logistic Support Force, PLA (Clinical Medical College of Fujian Medical University, Dongfang Hospital Affiliated to Xiamen University), Fuzhou 350025, Fujian Province, China
| | - Wei Chen
- Department of General Practice, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, Shaanxi Province, China
| | - Dongyu Wei
- Center of Clinical Aerospace Medicine, Fourth Military Medical University, Xi'an 710032, Shaanxi Province, China.,Department of Aviation Medicine, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, Shaanxi Province, China
| | - Siwang Wang
- Department of Chinese Material Medical and Natural Medicines, Fourth Military Medical University, Xi'an 710032, Shaanxi Province, China
| | - Zuoming Zhang
- Center of Clinical Aerospace Medicine, Fourth Military Medical University, Xi'an 710032, Shaanxi Province, China.,Department of Aviation Medicine, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, Shaanxi Province, China
| | - Wei Ge
- Department of General Practice, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, Shaanxi Province, China
| | - Tao Chen
- Center of Clinical Aerospace Medicine, Fourth Military Medical University, Xi'an 710032, Shaanxi Province, China.,Department of Aviation Medicine, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, Shaanxi Province, China
| |
Collapse
|
25
|
Lottes EN, Cox DN. Homeostatic Roles of the Proteostasis Network in Dendrites. Front Cell Neurosci 2020; 14:264. [PMID: 33013325 PMCID: PMC7461941 DOI: 10.3389/fncel.2020.00264] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Accepted: 07/28/2020] [Indexed: 12/13/2022] Open
Abstract
Cellular protein homeostasis, or proteostasis, is indispensable to the survival and function of all cells. Distinct from other cell types, neurons are long-lived, exhibiting architecturally complex and diverse multipolar projection morphologies that can span great distances. These properties present unique demands on proteostatic machinery to dynamically regulate the neuronal proteome in both space and time. Proteostasis is regulated by a distributed network of cellular processes, the proteostasis network (PN), which ensures precise control of protein synthesis, native conformational folding and maintenance, and protein turnover and degradation, collectively safeguarding proteome integrity both under homeostatic conditions and in the contexts of cellular stress, aging, and disease. Dendrites are equipped with distributed cellular machinery for protein synthesis and turnover, including dendritically trafficked ribosomes, chaperones, and autophagosomes. The PN can be subdivided into an adaptive network of three major functional pathways that synergistically govern protein quality control through the action of (1) protein synthesis machinery; (2) maintenance mechanisms including molecular chaperones involved in protein folding; and (3) degradative pathways (e.g., Ubiquitin-Proteasome System (UPS), endolysosomal pathway, and autophagy. Perturbations in any of the three arms of proteostasis can have dramatic effects on neurons, especially on their dendrites, which require tightly controlled homeostasis for proper development and maintenance. Moreover, the critical importance of the PN as a cell surveillance system against protein dyshomeostasis has been highlighted by extensive work demonstrating that the aggregation and/or failure to clear aggregated proteins figures centrally in many neurological disorders. While these studies demonstrate the relevance of derangements in proteostasis to human neurological disease, here we mainly review recent literature on homeostatic developmental roles the PN machinery plays in the establishment, maintenance, and plasticity of stable and dynamic dendritic arbors. Beyond basic housekeeping functions, we consider roles of PN machinery in protein quality control mechanisms linked to dendritic plasticity (e.g., dendritic spine remodeling during LTP); cell-type specificity; dendritic morphogenesis; and dendritic pruning.
Collapse
Affiliation(s)
| | - Daniel N. Cox
- Neuroscience Institute, Georgia State University, Atlanta, GA, United States
| |
Collapse
|
26
|
Martínez Corrales G, Alic N. Evolutionary Conservation of Transcription Factors Affecting Longevity. Trends Genet 2020; 36:373-382. [PMID: 32294417 DOI: 10.1016/j.tig.2020.02.003] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Revised: 02/12/2020] [Accepted: 02/18/2020] [Indexed: 12/29/2022]
Abstract
The increasing number of older people is resulting in an increased prevalence of age-related diseases. Research has shown that the ageing process itself is a potential point of intervention. Indeed, gene expression can be optimised for health in older ages through manipulation of transcription factor (TF) activity. This review is focused on the ever-growing number of TFs whose effects on ageing are evolutionarily conserved. These regulate a plethora of functions, including stress resistance, metabolism, and growth. They are engaged in complex interactions within and between different cell types, impacting the physiology of the entire organism. Since ageing is not programmed, the conservation of their effects on lifespan is most likely a reflection of the conservation of their functions in youth.
Collapse
|
27
|
Song JY, Wang XG, Zhang ZY, Che L, Fan B, Li GY. Endoplasmic reticulum stress and the protein degradation system in ophthalmic diseases. PeerJ 2020; 8:e8638. [PMID: 32117642 PMCID: PMC7036270 DOI: 10.7717/peerj.8638] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2019] [Accepted: 01/26/2020] [Indexed: 12/16/2022] Open
Abstract
Objective Endoplasmic reticulum (ER) stress is involved in the pathogenesis of various ophthalmic diseases, and ER stress-mediated degradation systems play an important role in maintaining ER homeostasis during ER stress. The purpose of this review is to explore the potential relationship between them and to find their equilibrium sites. Design This review illustrates the important role of reasonable regulation of the protein degradation system in ER stress-mediated ophthalmic diseases. There were 128 articles chosen for review in this study, and the keywords used for article research are ER stress, autophagy, UPS, ophthalmic disease, and ocular. Data sources The data are from Web of Science, PubMed, with no language restrictions from inception until 2019 Jul. Results The ubiquitin proteasome system (UPS) and autophagy are important degradation systems in ER stress. They can restore ER homeostasis, but if ER stress cannot be relieved in time, cell death may occur. However, they are not independent of each other, and the relationship between them is complementary. Therefore, we propose that ER stability can be achieved by adjusting the balance between them. Conclusion The degradation system of ER stress, UPS and autophagy are interrelated. Because an imbalance between the UPS and autophagy can cause cell death, regulating that balance may suppress ER stress and protect cells against pathological stress damage.
Collapse
Affiliation(s)
- Jing-Yao Song
- Department of Ophthalmology, Second Hospital of Jilin University, ChangChun, China
| | - Xue-Guang Wang
- Department of Traumatic Orthopedics, Third People's Hospital of Jinan, Jinan, China
| | - Zi-Yuan Zhang
- Department of Ophthalmology, Second Hospital of Jilin University, ChangChun, China
| | - Lin Che
- Department of Ophthalmology, Second Hospital of Jilin University, ChangChun, China
| | - Bin Fan
- Department of Ophthalmology, Second Hospital of Jilin University, ChangChun, China
| | - Guang-Yu Li
- Department of Ophthalmology, Second Hospital of Jilin University, ChangChun, China
| |
Collapse
|
28
|
Beneficial effect of ER stress preconditioning in protection against FFA-induced adipocyte inflammation via XBP1 in 3T3-L1 adipocytes. Mol Cell Biochem 2019; 463:45-55. [PMID: 31630283 PMCID: PMC6946732 DOI: 10.1007/s11010-019-03627-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2019] [Accepted: 09/12/2019] [Indexed: 01/03/2023]
Abstract
Adipose tissue inflammation is closely associated with the development of obesity and insulin resistance. Free fatty acids (FFAs) are a major inducer of obesity-related insulin resistance. Previously, we reported that endoplasmic reticulum (ER) stress potentially mediated retinal inflammation in diabetic retinopathy. The unfolded protein response (UPR) protects cells against damage induced by oxidative stress. X-box binding protein 1 (XBP1) plays a major role in protecting cells by modulating the UPR. However, the link between ER stress and adipocyte inflammation has been poorly investigated. In the present study, we found that pretreatment of 3T3-L1 adipocytes with a low dose of ER stress inducer tunicamycin inhibited FFA-induced upregulated expression of inflammatory cytokines. In addition, FFAs induced phosphorylation of the p65 subunit of NF-κB was largely inhibited by pretreatment with tunicamycin in 3T3-L1 adipocytes. Knockdown of XBP1 by siRNA markedly mitigated the protective effects of preconditioning against inflammation. Conversely, overexpression of XBP1 alleviated FFA-induced phosphorylation of IκB-α, IKKα/β, and NF-κB, which was accompanied by decreased inflammatory cytokine expression. Collectively, these results imply a beneficial role of ER stress preconditioning in protecting against FFA-induced 3T3-L1 adipocyte inflammation, which is likely mediated through inhibition of the IKK/NF-κB pathway via XBP1.
Collapse
|
29
|
McLaughlin T, Siddiqi M, Wang JJ, Zhang SX. Loss of XBP1 Leads to Early-Onset Retinal Neurodegeneration in a Mouse Model of Type I Diabetes. J Clin Med 2019; 8:jcm8060906. [PMID: 31242599 PMCID: PMC6617367 DOI: 10.3390/jcm8060906] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2019] [Revised: 06/18/2019] [Accepted: 06/21/2019] [Indexed: 12/18/2022] Open
Abstract
Retinal neuronal injury and degeneration is one of the primary manifestations of diabetic retinopathy, a leading cause of vision loss in working age adults. In pathological conditions, including diabetes and some physiological conditions such as aging, protein homeostasis can become disrupted, leading to endoplasmic reticulum (ER) stress. Severe or unmitigated ER stress can lead to cell death, which in retinal neurons results in irreversible loss of visual function. X-box binding protein 1 (XBP1) is a major transcription factor responsible for the adaptive unfolded protein response (UPR) to maintain protein homeostasis in cells undergoing ER stress. The purpose of this study is to determine the role of XBP1-mediated UPR in retinal neuronal survival and function in a mouse model of type 1 diabetes. Using a conditional retina-specific XBP1 knockout mouse line, we demonstrate that depletion of XBP1 in retinal neurons results in early onset retinal function decline, loss of retinal ganglion cells and photoreceptors, disrupted photoreceptor ribbon synapses, and Müller cell activation after induction of diabetes. Our findings suggest an important role of XBP1-mediated adaptive UPR in retinal neuronal survival and function in diabetes.
Collapse
Affiliation(s)
- Todd McLaughlin
- Departments of Ophthalmology and Ross Eye Institute, University at Buffalo, Buffalo, NY 14203, USA.
- SUNY Eye Institute, State University of New York, Buffalo, NY 14203, USA.
| | - Manhal Siddiqi
- Departments of Ophthalmology and Ross Eye Institute, University at Buffalo, Buffalo, NY 14203, USA.
- SUNY Eye Institute, State University of New York, Buffalo, NY 14203, USA.
| | - Joshua J Wang
- Departments of Ophthalmology and Ross Eye Institute, University at Buffalo, Buffalo, NY 14203, USA.
- SUNY Eye Institute, State University of New York, Buffalo, NY 14203, USA.
| | - Sarah X Zhang
- Departments of Ophthalmology and Ross Eye Institute, University at Buffalo, Buffalo, NY 14203, USA.
- SUNY Eye Institute, State University of New York, Buffalo, NY 14203, USA.
- Department of Biochemistry, State University of New York, Buffalo, NY 14203, USA.
| |
Collapse
|
30
|
McLaughlin T, Dhimal N, Li J, Wang JJ, Zhang SX. p58 IPK Is an Endogenous Neuroprotectant for Retinal Ganglion Cells. Front Aging Neurosci 2018; 10:267. [PMID: 30245625 PMCID: PMC6137320 DOI: 10.3389/fnagi.2018.00267] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2018] [Accepted: 08/20/2018] [Indexed: 11/13/2022] Open
Abstract
p58IPK is an endoplasmic reticulum (ER)-resident chaperone playing a critical role in facilitating protein folding and protein homeostasis. Previously, we have demonstrated that p58IPK is expressed broadly in retinal neurons including retinal ganglion cells (RGCs) and loss of p58IPK results in age-related RGC degeneration. In the present study, we investigate the role of p58IPK in neuroprotection by in vitro and in vivo studies using primary RGC culture and two well-established disease-relevant RGC injury models: retinal ischemia/reperfusion (I/R) and microbead-induced ocular hypertension. Our results demonstrate that in both in vivo models, p58IPK -/- mice exhibit significantly increased RGC loss compared to wild type (WT) mice. In vitro, p58IPK-deficient RGCs show reduced viability and are more susceptible to cell death induced by the ER stress inducer tunicamycin (TM). Overexpression of p58IPK by adeno-associated virus (AAV) significantly diminishes TM-induced cell death in both WT and p58IPK -/- RGCs. Interestingly, we find that loss of p58IPK leads to reduced mRNA expression, but not the protein level, of mesencephalic astrocyte-derived neurotrophic factor (MANF), a neurotrophic factor that resides in the ER. Treatment with recombinant MANF protein protects R28 retinal neural cells and mouse retinal explants from TM-induced cell death. Taken together, our study suggests that p58IPK functions as an endogenous neuroprotectant for RGCs. The mechanisms underlying p58IPK's neuroprotective action and the potential interactions between p58IPK and MANF warrant future investigation.
Collapse
Affiliation(s)
- Todd McLaughlin
- Departments of Ophthalmology and Biochemistry, Ross Eye Institute, University at Buffalo, State University of New York, Buffalo, NY, United States.,SUNY Eye Institute, State University of New York, Buffalo, NY, United States
| | - Narayan Dhimal
- Departments of Ophthalmology and Biochemistry, Ross Eye Institute, University at Buffalo, State University of New York, Buffalo, NY, United States
| | - Junhua Li
- Departments of Ophthalmology and Biochemistry, Ross Eye Institute, University at Buffalo, State University of New York, Buffalo, NY, United States
| | - Joshua Jianxin Wang
- Departments of Ophthalmology and Biochemistry, Ross Eye Institute, University at Buffalo, State University of New York, Buffalo, NY, United States.,SUNY Eye Institute, State University of New York, Buffalo, NY, United States
| | - Sarah Xin Zhang
- Departments of Ophthalmology and Biochemistry, Ross Eye Institute, University at Buffalo, State University of New York, Buffalo, NY, United States.,SUNY Eye Institute, State University of New York, Buffalo, NY, United States
| |
Collapse
|
31
|
Kelly K, Wang JJ, Zhang SX. The unfolded protein response signaling and retinal Müller cell metabolism. Neural Regen Res 2018; 13:1861-1870. [PMID: 30233053 PMCID: PMC6183030 DOI: 10.4103/1673-5374.239431] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
The retina is one of the most energy demanding tissues in the body. Like most neurons in the central nervous system, retinal neurons consume high amounts of adenosine-5'-triphosphate (ATP) to generate visual signal and transmit the information to the brain. Disruptions in retinal metabolism can cause neuronal dysfunction and degeneration resulting in severe visual impairment and even blindness. The homeostasis of retinal metabolism is tightly controlled by multiple signaling pathways, such as the unfolded protein response (UPR), and the close interactions between retinal neurons and other retinal cell types including vascular cells and Müller glia. The UPR is a highly conserved adaptive cellular response and can be triggered by many physiological stressors and pathophysiological conditions. Activation of the UPR leads to changes in glycolytic rate, ATP production, de novo serine synthesis, and the hexosamine biosynthetic pathway, which are considered critical components of Müller glia metabolism and provide metabolic support to surrounding neurons. When these pathways are disrupted, neurodegeneration occurs rapidly. In this review, we summarize recent advance in studies of the UPR in Müller glia and highlight the potential role of the UPR in retinal degeneration through regulation of Müller glia metabolism.
Collapse
Affiliation(s)
- Kristen Kelly
- Department of Ophthalmology and Neuroscience Program, Ross Eye Institute, University at Buffalo, State University of New York, Buffalo, NY, USA
| | - Joshua J Wang
- Department of Ophthalmology and Neuroscience Program, Ross Eye Institute, University at Buffalo; SUNY Eye Institute, State University of New York, Buffalo, NY, USA
| | - Sarah X Zhang
- Department of Ophthalmology and Neuroscience Program, Ross Eye Institute, University at Buffalo; SUNY Eye Institute, State University of New York, Buffalo, NY, USA
| |
Collapse
|