1
|
Tang S, Yang J, Xiao B, Wang Y, Lei Y, Lai D, Qiu Q. Aberrant Lipid Metabolism and Complement Activation in Age-Related Macular Degeneration. Invest Ophthalmol Vis Sci 2024; 65:20. [PMID: 39405051 PMCID: PMC11482642 DOI: 10.1167/iovs.65.12.20] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Accepted: 09/10/2024] [Indexed: 10/19/2024] Open
Abstract
Age-related macular degeneration (AMD) stands as a leading cause of severe visual impairment and blindness among the elderly globally. As a multifactorial disease, AMD's pathogenesis is influenced by genetic, environmental, and age-related factors, with lipid metabolism abnormalities and complement system dysregulation playing critical roles. This review delves into recent advancements in understanding the intricate interaction between these two crucial pathways, highlighting their contribution to the disease's progression through chronic inflammation, drusen formation, and retinal pigment epithelium dysfunction. Importantly, emerging evidence points to dysregulated lipid profiles, particularly alterations in high-density lipoprotein levels, oxidized lipid deposits, and intracellular lipofuscin accumulation, as exacerbating factors that enhance complement activation and subsequently amplify tissue damage in AMD. Furthermore, genetic studies have revealed significant associations between AMD and specific genes involved in lipid transport and complement regulation, shedding light on disease susceptibility and underlying mechanisms. The review further explores the clinical implications of these findings, advocating for a novel therapeutic approach that integrates lipid metabolism modulators with complement inhibitors. By concurrently targeting these pathways, the dual-targeted approach holds promise in significantly improving outcomes for AMD patients, heralding a new horizon in AMD management and treatment.
Collapse
Affiliation(s)
- Siao Tang
- Xiangya School of Medicine, Central South University, Changsha, Hunan, PR China
- Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, PR China
- National Clinical Research Center for Eye Diseases, Shanghai Key Laboratory of Ocular Fundus Diseases, Shanghai Engineering Center for Visual Science and Photomedicine, Shanghai Engineering Center for Precise Diagnosis and Treatment of Eye Diseases, Shanghai, PR China
| | - Jiaqi Yang
- Xiangya School of Medicine, Central South University, Changsha, Hunan, PR China
- Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, PR China
- National Clinical Research Center for Eye Diseases, Shanghai Key Laboratory of Ocular Fundus Diseases, Shanghai Engineering Center for Visual Science and Photomedicine, Shanghai Engineering Center for Precise Diagnosis and Treatment of Eye Diseases, Shanghai, PR China
| | - Bingqing Xiao
- Xiangya School of Medicine, Central South University, Changsha, Hunan, PR China
- Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, PR China
- National Clinical Research Center for Eye Diseases, Shanghai Key Laboratory of Ocular Fundus Diseases, Shanghai Engineering Center for Visual Science and Photomedicine, Shanghai Engineering Center for Precise Diagnosis and Treatment of Eye Diseases, Shanghai, PR China
| | - Yani Wang
- Xiangya School of Medicine, Central South University, Changsha, Hunan, PR China
- Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, PR China
- National Clinical Research Center for Eye Diseases, Shanghai Key Laboratory of Ocular Fundus Diseases, Shanghai Engineering Center for Visual Science and Photomedicine, Shanghai Engineering Center for Precise Diagnosis and Treatment of Eye Diseases, Shanghai, PR China
| | - Yiou Lei
- Xiangya School of Medicine, Central South University, Changsha, Hunan, PR China
- Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, PR China
- National Clinical Research Center for Eye Diseases, Shanghai Key Laboratory of Ocular Fundus Diseases, Shanghai Engineering Center for Visual Science and Photomedicine, Shanghai Engineering Center for Precise Diagnosis and Treatment of Eye Diseases, Shanghai, PR China
| | - Dongwei Lai
- Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, PR China
- National Clinical Research Center for Eye Diseases, Shanghai Key Laboratory of Ocular Fundus Diseases, Shanghai Engineering Center for Visual Science and Photomedicine, Shanghai Engineering Center for Precise Diagnosis and Treatment of Eye Diseases, Shanghai, PR China
| | - Qinghua Qiu
- Department of Ophthalmology, Tong Ren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, PR China
| |
Collapse
|
2
|
Alic L, Binder CJ, Papac-Milicevic N. The OSE complotype and its clinical potential. Front Immunol 2022; 13:1010893. [PMID: 36248824 PMCID: PMC9561429 DOI: 10.3389/fimmu.2022.1010893] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Accepted: 09/12/2022] [Indexed: 11/17/2022] Open
Abstract
Cellular death, aging, and tissue damage trigger inflammation that leads to enzymatic and non-enzymatic lipid peroxidation of polyunsaturated fatty acids present on cellular membranes and lipoproteins. This results in the generation of highly reactive degradation products, such as malondialdehyde (MDA) and 4-hydroxynonenal (4-HNE), that covalently modify free amino groups of proteins and lipids in their vicinity. These newly generated neoepitopes represent a unique set of damage-associated molecular patterns (DAMPs) associated with oxidative stress termed oxidation-specific epitopes (OSEs). OSEs are enriched on oxidized lipoproteins, microvesicles, and dying cells, and can trigger sterile inflammation. Therefore, prompt recognition and removal of OSEs is required to maintain the homeostatic balance. This is partially achieved by various humoral components of the innate immune system, such as natural IgM antibodies, pentraxins and complement components that not only bind OSEs but in some cases modulate their pro-inflammatory potential. Natural IgM antibodies are potent complement activators, and 30% of them recognize OSEs such as oxidized phosphocholine (OxPC-), 4-HNE-, and MDA-epitopes. Furthermore, OxPC-epitopes can bind the complement-activating pentraxin C-reactive protein, while MDA-epitopes are bound by C1q, C3a, complement factor H (CFH), and complement factor H-related proteins 1, 3, 5 (FHR-1, FHR-3, FHR-5). In addition, CFH and FHR-3 are recruited to 2-(ω-carboxyethyl)pyrrole (CEP), and full-length CFH also possesses the ability to attenuate 4-HNE-induced oxidative stress. Consequently, alterations in the innate humoral defense against OSEs predispose to the development of diseases associated with oxidative stress, as shown for the prototypical OSE, MDA-epitopes. In this mini-review, we focus on the mechanisms of the accumulation of OSEs, the pathophysiological consequences, and the interactions between different OSEs and complement components. Additionally, we will discuss the clinical potential of genetic variants in OSE-recognizing complement proteins – the OSE complotype - in the risk estimation of diseases associated with oxidative stress.
Collapse
Affiliation(s)
- Lejla Alic
- Department of Medical Biochemistry, Faculty of Medicine, University of Sarajevo, Sarajevo, Bosnia and Herzegovina
| | - Christoph J. Binder
- Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria
| | - Nikolina Papac-Milicevic
- Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria
- *Correspondence: Nikolina Papac-Milicevic,
| |
Collapse
|
3
|
Tolentino MJ, Tolentino AJ. Investigational drugs in clinical trials for macular degeneration. Expert Opin Investig Drugs 2022; 31:1067-1085. [PMID: 35962560 DOI: 10.1080/13543784.2022.2113375] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
INTRODUCTION Intravitreal anti-vascular endothelial growth factor (VEGF) injections for exudative age-related macular degeneration (eAMD) are effective and safe but require frequent injections and have nonresponding patients. Geographic atrophy/dry AMD (gaAMD) remains an unmet medical need . New therapies are needed to address this leading cause of blindness in the increasing aged population. AREAS COVERED This paper reviews the pathogenesis of macular degeneration, current and failed therapeutics, therapies undergoing clinical trials and a rationale for why certain AMD therapies may succeed or fail . EXPERT OPINION VEGF- inhibitors reduce both vascular leakage and neovascularization. Experimental therapies that only address neovascularization or leakage will unlikely supplant anti-VEGF therapies. The most promising future therapies for eAMD, are those that target, more potently inhibit and have a more sustained effect on the VEGF pathway such as KSI-301, RGX-314, CLS-AX, EYEP-1901, OTX-TKI. GaAMD is a phenotype of phagocytic retinal cell loss. Inhibiting phagocytic activity of retinal microglial/macrophages at the border of GA and reducing complement derived activators of microglial/macrophage is the most promising strategy. Complement inhibitors (Pegcetacoplan and Avacincaptad pegol) will likely obtain FDA approval but will serve to pave the way for combined complement and direct phagocytic inhibitors such as AVD-104.
Collapse
Affiliation(s)
- Michael J Tolentino
- University of Central Florida, FL, USA.,Blue Ocean Clinical Research, Lakeland, FL, USA.,Aviceda Therapeutics, Cambridge, MA, USA
| | | |
Collapse
|
4
|
Lewandowski D, Sander CL, Tworak A, Gao F, Xu Q, Skowronska-Krawczyk D. Dynamic lipid turnover in photoreceptors and retinal pigment epithelium throughout life. Prog Retin Eye Res 2022; 89:101037. [PMID: 34971765 PMCID: PMC10361839 DOI: 10.1016/j.preteyeres.2021.101037] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Revised: 12/13/2021] [Accepted: 12/15/2021] [Indexed: 12/13/2022]
Abstract
The retinal pigment epithelium-photoreceptor interphase is renewed each day in a stunning display of cellular interdependence. While photoreceptors use photosensitive pigments to convert light into electrical signals, the RPE supports photoreceptors in their function by phagocytizing shed photoreceptor tips, regulating the blood retina barrier, and modulating inflammatory responses, as well as regenerating the 11-cis-retinal chromophore via the classical visual cycle. These processes involve multiple protein complexes, tightly regulated ligand-receptors interactions, and a plethora of lipids and protein-lipids interactions. The role of lipids in maintaining a healthy interplay between the RPE and photoreceptors has not been fully delineated. In recent years, novel technologies have resulted in major advancements in understanding several facets of this interplay, including the involvement of lipids in phagocytosis and phagolysosome function, nutrient recycling, and the metabolic dependence between the two cell types. In this review, we aim to integrate the complex role of lipids in photoreceptor and RPE function, emphasizing the dynamic exchange between the cells as well as discuss how these processes are affected in aging and retinal diseases.
Collapse
Affiliation(s)
- Dominik Lewandowski
- Department of Ophthalmology, Center for Translational Vision Research, School of Medicine, UC Irvine, Irvine, CA, USA
| | - Christopher L Sander
- Department of Ophthalmology, Center for Translational Vision Research, School of Medicine, UC Irvine, Irvine, CA, USA; Department of Pharmacology, School of Medicine, Case Western Reserve University, Cleveland, OH, USA
| | - Aleksander Tworak
- Department of Ophthalmology, Center for Translational Vision Research, School of Medicine, UC Irvine, Irvine, CA, USA
| | - Fangyuan Gao
- Department of Ophthalmology, Center for Translational Vision Research, School of Medicine, UC Irvine, Irvine, CA, USA
| | - Qianlan Xu
- Department of Physiology and Biophysics, Center for Translational Vision Research, School of Medicine, UC Irvine, Irvine, CA, USA
| | - Dorota Skowronska-Krawczyk
- Department of Ophthalmology, Center for Translational Vision Research, School of Medicine, UC Irvine, Irvine, CA, USA; Department of Physiology and Biophysics, Center for Translational Vision Research, School of Medicine, UC Irvine, Irvine, CA, USA.
| |
Collapse
|
5
|
Chu-Tan JA, Cioanca AV, Feng ZP, Wooff Y, Schumann U, Aggio-Bruce R, Patel H, Rutar M, Hannan K, Panov K, Provis J, Natoli R. Functional microRNA targetome undergoes degeneration-induced shift in the retina. Mol Neurodegener 2021; 16:60. [PMID: 34465369 PMCID: PMC8406976 DOI: 10.1186/s13024-021-00478-9] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Accepted: 08/03/2021] [Indexed: 01/05/2023] Open
Abstract
BACKGROUND MicroRNA (miRNA) play a significant role in the pathogenesis of complex neurodegenerative diseases including age-related macular degeneration (AMD), acting as post-transcriptional gene suppressors through their association with argonaute 2 (AGO2) - a key member of the RNA Induced Silencing Complex (RISC). Identifying the retinal miRNA/mRNA interactions in health and disease will provide important insight into the key pathways miRNA regulate in disease pathogenesis and may lead to potential therapeutic targets to mediate retinal degeneration. METHODS To identify the active miRnome targetome interactions in the healthy and degenerating retina, AGO2 HITS-CLIP was performed using a rodent model of photoreceptor degeneration. Analysis of publicly available single-cell RNA sequencing (scRNAseq) data was performed to identify the cellular location of AGO2 and key members of the microRNA targetome in the retina. AGO2 findings were verified by in situ hybridization (RNA) and immunohistochemistry (protein). RESULTS Analysis revealed a similar miRnome between healthy and damaged retinas, however, a shift in the active targetome was observed with an enrichment of miRNA involvement in inflammatory pathways. This shift was further demonstrated by a change in the seed binding regions of miR-124-3p, the most abundant retinal AGO2-bound miRNA, and has known roles in regulating retinal inflammation. Additionally, photoreceptor cluster miR-183/96/182 were all among the most highly abundant miRNA bound to AGO2. Following damage, AGO2 expression was localized to the inner retinal layers and more in the OLM than in healthy retinas, indicating a locational miRNA response to retinal damage. CONCLUSIONS This study provides important insight into the alteration of miRNA regulatory activity that occurs as a response to retinal degeneration and explores the miRNA-mRNA targetome as a consequence of retinal degenerations. Further characterisation of these miRNA/mRNA interactions in the context of the degenerating retina may provide an important insight into the active role these miRNA may play in diseases such as AMD.
Collapse
Affiliation(s)
- Joshua A. Chu-Tan
- Eccles Institute of Neuroscience, The John Curtin School of Medical Research, College of Health and Medicine, The Australian National University, Acton, Canberra, ACT 2601 Australia
- The Australian National University Medical School, College of Health and Medicine, Canberra, ACT 2601 Australia
| | - Adrian V. Cioanca
- Eccles Institute of Neuroscience, The John Curtin School of Medical Research, College of Health and Medicine, The Australian National University, Acton, Canberra, ACT 2601 Australia
| | - Zhi-Ping Feng
- The ANU Bioinformatics Consultancy, The John Curtin School of Medical Research, College of Health and Medicine, The Australian National University, Acton, Canberra, ACT 2601 Australia
| | - Yvette Wooff
- Eccles Institute of Neuroscience, The John Curtin School of Medical Research, College of Health and Medicine, The Australian National University, Acton, Canberra, ACT 2601 Australia
- The Australian National University Medical School, College of Health and Medicine, Canberra, ACT 2601 Australia
| | - Ulrike Schumann
- Eccles Institute of Neuroscience, The John Curtin School of Medical Research, College of Health and Medicine, The Australian National University, Acton, Canberra, ACT 2601 Australia
| | - Riemke Aggio-Bruce
- Eccles Institute of Neuroscience, The John Curtin School of Medical Research, College of Health and Medicine, The Australian National University, Acton, Canberra, ACT 2601 Australia
- The Australian National University Medical School, College of Health and Medicine, Canberra, ACT 2601 Australia
| | - Hardip Patel
- The ANU Bioinformatics Consultancy, The John Curtin School of Medical Research, College of Health and Medicine, The Australian National University, Acton, Canberra, ACT 2601 Australia
| | - Matt Rutar
- School of Biomedical Sciences, The University of Melbourne, Parkville, Victoria 3010 Australia
- Faculty of Science and Technology, University of Canberra, Bruce, ACT 2617 Australia
| | - Katherine Hannan
- ACRF Department of Cancer Biology and Therapeutics, The John Curtin School of Medical Research, College of Health and Medicine, The Australian National University, Acton, Canberra, ACT 2601 Australia
| | - Konstantin Panov
- School of Biological Sciences Queen’s University Belfast, Belfast, BT9 5DL Northern Ireland
| | - Jan Provis
- Eccles Institute of Neuroscience, The John Curtin School of Medical Research, College of Health and Medicine, The Australian National University, Acton, Canberra, ACT 2601 Australia
- The Australian National University Medical School, College of Health and Medicine, Canberra, ACT 2601 Australia
| | - Riccardo Natoli
- Eccles Institute of Neuroscience, The John Curtin School of Medical Research, College of Health and Medicine, The Australian National University, Acton, Canberra, ACT 2601 Australia
- The Australian National University Medical School, College of Health and Medicine, Canberra, ACT 2601 Australia
| |
Collapse
|
6
|
Fedintsev A, Moskalev A. Stochastic non-enzymatic modification of long-lived macromolecules - A missing hallmark of aging. Ageing Res Rev 2020; 62:101097. [PMID: 32540391 DOI: 10.1016/j.arr.2020.101097] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Revised: 05/05/2020] [Accepted: 06/04/2020] [Indexed: 12/12/2022]
Abstract
Damage accumulation in long-living macromolecules (especially extracellular matrix (ECM) proteins, nuclear pore complex (NPC) proteins, and histones) is a missing hallmark of aging. Stochastic non-enzymatic modifications of ECM trigger cellular senescence as well as many other hallmarks of aging affect organ barriers integrity and drive tissue fibrosis. The importance of it for aging makes it a key target for interventions. The most promising of them can be AGE inhibitors (chelators, O-acetyl group or transglycating activity compounds, amadorins and amadoriases), glucosepane breakers, stimulators of elastogenesis, and RAGE antagonists.
Collapse
Affiliation(s)
- Alexander Fedintsev
- Institute of Biology of FRC of Komi Scientific Center, Ural Branch of Russian Academy of Sciences, Syktyvkar, Russia
| | - Alexey Moskalev
- Institute of Biology of FRC of Komi Scientific Center, Ural Branch of Russian Academy of Sciences, Syktyvkar, Russia.
| |
Collapse
|
7
|
Will-Orrego A, Qiu Y, Fassbender ES, Shen S, Aranda J, Kotagiri N, Maker M, Liao SM, Jaffee BD, Poor SH. Amount of Mononuclear Phagocyte Infiltrate Does Not Predict Area of Experimental Choroidal Neovascularization (CNV). J Ocul Pharmacol Ther 2019; 34:489-499. [PMID: 30188257 PMCID: PMC6152860 DOI: 10.1089/jop.2017.0131] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Purpose: Mononuclear phagocytes (MNPs) are present in neovascular age-related macular degeneration (nv AMD) which is also called choroidal neovascularization (CNV). The number and phenotype of the MNPs depend upon the local environment in the CNV and effect of nv AMD therapy. We investigated ocular cell infiltration and conditions that modulate angiogenesis in a laser-induced mouse CNV model. Methods: We developed assays to quantify MNPs in our established mouse CNV model. One MNP assay quantified the number of subretinal cells peripheral to the CNV lesions. A second assay semiquantitatively assesses the number of MNPs localized to the CNV lesion. We used these assays to measure the effect of toll-like receptor-2 (TLR-2) activation, anti-vascular endothelial growth factor (VEGF) therapy, and chemokine (C-C motif) ligand 2 (Ccl2) genetic deletion on MNP infiltration after laser injury. Results: Laser injury induced blood vessel growth and infiltration of MNPs. Systemic administration of a TLR-2 activating peptide increased laser-induced CNV area, MNP cell numbers, and MNP density over the CNV lesions. Systemic administration of a VEGF antibody reduced CNV area, while Ccl2 genetic deletion increased CNV area. Despite the change in amount of angiogenesis, MNP infiltration was, surprisingly, unchanged in these 2 conditions. Conclusions: MNP quantification provides biological insights for candidate AMD therapies. The number of infiltrating MNP cells does not correlate with the amount of laser-induced CNV area.
Collapse
Affiliation(s)
- Adrian Will-Orrego
- Department of Ophthalmology, Novartis Institutes for Biomedical Research , Cambridge, Massachusetts
| | - Yubin Qiu
- Department of Ophthalmology, Novartis Institutes for Biomedical Research , Cambridge, Massachusetts
| | - Elizabeth S Fassbender
- Department of Ophthalmology, Novartis Institutes for Biomedical Research , Cambridge, Massachusetts
| | - Siyuan Shen
- Department of Ophthalmology, Novartis Institutes for Biomedical Research , Cambridge, Massachusetts
| | - Jorge Aranda
- Department of Ophthalmology, Novartis Institutes for Biomedical Research , Cambridge, Massachusetts
| | - Namrata Kotagiri
- Department of Ophthalmology, Novartis Institutes for Biomedical Research , Cambridge, Massachusetts
| | - Michael Maker
- Department of Ophthalmology, Novartis Institutes for Biomedical Research , Cambridge, Massachusetts
| | - Sha-Mei Liao
- Department of Ophthalmology, Novartis Institutes for Biomedical Research , Cambridge, Massachusetts
| | - Bruce D Jaffee
- Department of Ophthalmology, Novartis Institutes for Biomedical Research , Cambridge, Massachusetts
| | - Stephen H Poor
- Department of Ophthalmology, Novartis Institutes for Biomedical Research , Cambridge, Massachusetts
| |
Collapse
|
8
|
Hou L, Yang G, Tang S, Alcazar C, Joshi P, Strassberg Z, Kim M, Kawamura M, Woo YJ, Shrager J, Ding S, Huang NF. Small Molecule Derived From Carboxyethylpyrrole Protein Adducts Promotes Angiogenesis in a Mouse Model of Peripheral Arterial Disease. J Am Heart Assoc 2019; 7:e009234. [PMID: 30371212 PMCID: PMC6222956 DOI: 10.1161/jaha.118.009234] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Background CEP (ω-[2-carboxyethyl]pyrrole) protein adducts are the end products of lipid oxidation associated with inflammation and have been implicated in the induction of angiogenesis in pathological conditions such as tissue ischemia. We synthesized small molecules derived from CEP protein adducts and evaluated the angiogenic effect of the CEP analog CEP 03 in the setting of peripheral arterial disease. Methods and Results The angiogenic effect of CEP 03 was assessed by in vitro analysis of primary human microvascular endothelial cell proliferation and tubelike formation in Matrigel (Corning). In the presence of CEP 03, proliferation of endothelial cells in vitro increased by 27±18% under hypoxic (1% O2) conditions, reaching similar levels to that of VEGF A (vascular endothelial growth factor A) stimulation (22±10%), relative to the vehicle control treatment. A similar effect of CEP 03 was demonstrated in the increased number of tubelike branches in Matrigel, reaching >70% induction in hypoxia, compared with the vehicle control. The therapeutic potential of CEP 03 was further evaluated in a mouse model of peripheral arterial disease by quantification of blood perfusion recovery and capillary density. In the ischemic hind limb, treatment of CEP 03 encapsulated within Matrigel significantly enhanced blood perfusion by 2-fold after 14 days compared with those treated with Matrigel alone. Moreover, these results concurred with histological finding that treatment of CEP 03 in Matrigel resulted in a significant increase in microvessel density compared with Matrigel alone. Conclusions Our data suggest that CEP 03 has a profound positive effect on angiogenesis and neovessel formation and thus has therapeutic potential for treatment of peripheral arterial disease.
Collapse
Affiliation(s)
- Luqia Hou
- 1 Stanford Cardiovascular Institute Stanford University Stanford CA.,2 Veterans Affairs Palo Alto Health Care System Palo Alto CA
| | - Guang Yang
- 1 Stanford Cardiovascular Institute Stanford University Stanford CA.,2 Veterans Affairs Palo Alto Health Care System Palo Alto CA
| | - Shibing Tang
- 3 Gladstone Institute of Cardiovascular Disease University of California San Francisco CA
| | - Cynthia Alcazar
- 1 Stanford Cardiovascular Institute Stanford University Stanford CA.,2 Veterans Affairs Palo Alto Health Care System Palo Alto CA
| | - Prajakta Joshi
- 1 Stanford Cardiovascular Institute Stanford University Stanford CA.,2 Veterans Affairs Palo Alto Health Care System Palo Alto CA
| | - Zachary Strassberg
- 1 Stanford Cardiovascular Institute Stanford University Stanford CA.,2 Veterans Affairs Palo Alto Health Care System Palo Alto CA
| | - Michael Kim
- 2 Veterans Affairs Palo Alto Health Care System Palo Alto CA.,4 Department of Cardiothoracic Surgery Stanford University Stanford CA
| | - Masashi Kawamura
- 1 Stanford Cardiovascular Institute Stanford University Stanford CA.,4 Department of Cardiothoracic Surgery Stanford University Stanford CA
| | - Y Joseph Woo
- 1 Stanford Cardiovascular Institute Stanford University Stanford CA.,4 Department of Cardiothoracic Surgery Stanford University Stanford CA
| | - Joseph Shrager
- 2 Veterans Affairs Palo Alto Health Care System Palo Alto CA.,4 Department of Cardiothoracic Surgery Stanford University Stanford CA
| | - Sheng Ding
- 3 Gladstone Institute of Cardiovascular Disease University of California San Francisco CA.,5 Department of Pharmaceutical Chemistry University of California San Francisco CA
| | - Ngan F Huang
- 1 Stanford Cardiovascular Institute Stanford University Stanford CA.,2 Veterans Affairs Palo Alto Health Care System Palo Alto CA.,4 Department of Cardiothoracic Surgery Stanford University Stanford CA
| |
Collapse
|
9
|
CFH exerts anti-oxidant effects on retinal pigment epithelial cells independently from protecting against membrane attack complex. Sci Rep 2019; 9:13873. [PMID: 31554875 PMCID: PMC6761137 DOI: 10.1038/s41598-019-50420-9] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2019] [Accepted: 08/20/2019] [Indexed: 12/03/2022] Open
Abstract
Age Related Macular Degeneration (AMD) is the first cause of social blindness in people aged over 65 leading to atrophy of retinal pigment epithelial cells (RPE), photoreceptors and choroids, eventually associated with choroidal neovascularization. Accumulation of undigested cellular debris within RPE cells or under the RPE (Drusen), oxidative stress and inflammatory mediators contribute to the RPE cell death. The major risk to develop AMD is the Y402H polymorphism of complement factor H (CFH). CFH interacting with oxidized phospholipids on the RPE membrane modulates the functions of these cells, but the exact role of CFH in RPE cell death and survival remain poorly understood. The aim of this study was to analyze the potential protective mechanism of CFH on RPE cells submitted to oxidative stress. Upon exposure to oxidized lipids 4-HNE (4-hydroxy-2-nonenal) derived from photoreceptors, both the human RPE cell line ARPE-19 and RPE cells derived from human induced pluripotent stem cells were protected from death only in the presence of the full length human recombinant CFH in the culture medium. This protective effect was independent from the membrane attack complex (MAC) formation. CFH maintained RPE cells tight junctions’ structure and regulated the caspase dependent apoptosis process. These results demonstrated the CFH anti-oxidative stress functions independently of its capacity to inhibit MAC formation.
Collapse
|
10
|
Huang YJ, Nan GX. Oxidative stress-induced angiogenesis. J Clin Neurosci 2019; 63:13-16. [DOI: 10.1016/j.jocn.2019.02.019] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2018] [Revised: 01/25/2019] [Accepted: 02/20/2019] [Indexed: 01/16/2023]
|
11
|
Damage-associated molecular pattern recognition is required for induction of retinal neuroprotective pathways in a sex-dependent manner. Sci Rep 2018; 8:9115. [PMID: 29904087 PMCID: PMC6002365 DOI: 10.1038/s41598-018-27479-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2018] [Accepted: 06/04/2018] [Indexed: 12/29/2022] Open
Abstract
Retinal degeneration is a common cause of irreversible blindness and is caused by the death of retinal light-sensitive neurons called photoreceptors. At the onset of degeneration, stressed photoreceptors cause retinal glial cells to secrete neuroprotective factors that slow the pace of degeneration. Leukemia inhibitory factor (LIF) is one such factor that is required for endogenous neuroprotection. Photoreceptors are known to release signals of cellular stress, called damage-associated molecular patterns (DAMPs) early in degeneration, and we hypothesized that receptors for DAMPs or pattern recognition receptors (PRRs) play a key role in the induction of LIF and neuroprotective stress responses in retinal glial cells. Toll-like receptor 2 (TLR2) is a well-established DAMP receptor. In our experiments, activation of TLR2 protected both male and female mice from light damage, while the loss of TLR2 in female mice did not impact photoreceptor survival. In contrast, induction of protective stress responses, microglial phenotype and photoreceptor survival were strongly impacted in male TLR2−/− mice. Lastly, using publicly available gene expression data, we show that TLR2 is expressed highly in resting microglia prior to injury, but is also induced in Müller cells in inherited retinal degeneration.
Collapse
|
12
|
Xu Q, Cao S, Rajapakse S, Matsubara JA. Understanding AMD by analogy: systematic review of lipid-related common pathogenic mechanisms in AMD, AD, AS and GN. Lipids Health Dis 2018; 17:3. [PMID: 29301530 PMCID: PMC5755337 DOI: 10.1186/s12944-017-0647-7] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2017] [Accepted: 12/17/2017] [Indexed: 12/15/2022] Open
Abstract
RATIONALE Age-related macular degeneration (AMD) is one of the leading causes of blindness among the elderly. Due to its complex etiology, current treatments have been insufficient. Previous studies reveal three systems closely involved in AMD pathogenesis: lipid metabolism, oxidation and inflammation. These systems are also involved in Alzheimer's disease, atherosclerosis and glomerulonephritis. Understanding commonalities of these four diseases may provide insight into AMD etiology. OBJECTIVES To understand AMD pathogenesis by analogy and suggest ideas for future research, this study summarizes main commonalities in disease pathogenesis of AMD, Alzheimer's disease, atherosclerosis and glomerulonephritis. METHODS Articles were identified through PubMed, Ovid Medline and Google Scholar. We summarized the common findings and synthesized critical differences. RESULTS Oxidation, lipid deposition, complement activation, and macrophage recruitment are involved in all four diseases shown by genetic, molecular, animal and human studies. Shared genetic variations further strengthen their connection. Potential areas for future research are suggested throughout the review. CONCLUSIONS The four diseases share many steps of an overall framework of pathogenesis. Various oxidative sources cause oxidative stress. Oxidized lipids and related molecules accumulate and lead to complement activation, macrophage recruitment and pathology. Investigations that arise under this structure may aid us to better understand AMD pathology.
Collapse
Affiliation(s)
- Qinyuan Xu
- Department of Ophthalmology and Visual Sciences, Faculty of Medicine, University of British Columbia, Vancouver, BC V5Z 3N9 Canada
| | - Sijia Cao
- Department of Ophthalmology and Visual Sciences, Faculty of Medicine, University of British Columbia, Vancouver, BC V5Z 3N9 Canada
| | - Sanjeeva Rajapakse
- Department of Ophthalmology and Visual Sciences, Faculty of Medicine, University of British Columbia, Vancouver, BC V5Z 3N9 Canada
| | - Joanne A. Matsubara
- Department of Ophthalmology and Visual Sciences, Faculty of Medicine, University of British Columbia, Vancouver, BC V5Z 3N9 Canada
| |
Collapse
|
13
|
Chistiakov DA, Melnichenko AA, Myasoedova VA, Grechko AV, Orekhov AN. Role of lipids and intraplaque hypoxia in the formation of neovascularization in atherosclerosis. Ann Med 2017; 49:661-677. [PMID: 28797175 DOI: 10.1080/07853890.2017.1366041] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
According to the current paradigm, chronic vascular inflammation plays a central role in the pathogenesis of atherosclerosis. The plaque progression is typically completed with rupture and subsequent acute cardiovascular complications. Previously, the role of adventitial vasa vasorum in atherogenesis was underestimated. However, investigators then revealed that vasa vasorum neovascularization can be observed when no clinical manifestation of atherosclerosis is present. Vasa vasorum is involved in various proatherogenic processes such as intimal accumulation of inflammatory leukocytes, intimal thickening, necrotic core formation, intraplaque haemorrhage, lesion rupture and atherothrombosis. Due to the destabilizing action of the intraplaque microenvironment, lesional vasa vasorum neovessels experience serious defects and abnormalities during development that leads to their immaturity, fragility and leakage. Indeed, intraplaque neovessels are a main cause of intraplaque haemorrhage. Visualization techniques showed that presence of neovascularization/haemorrhage can serve as a good indicator of lesion instability and higher risk of rupture. Vasa vasorum density is a strong predictor of acute cardiovascular events such as sudden death, myocardial infarction and stroke. At present, arterial vasa vasorum neovascularization is under intensive investigation along with development of therapeutic tools focused on the control of formation of vasa vasorum neovessels in order to prevent plaque haemorrhage/rupture and thromboembolism. KEY MESSAGE Neovascularization plays an important role in atherosclerosis, being involved in unstable plaque formation. Presence of neovascularization and haemorrhage indicates plaque instability and risk of rupture. Various imaging techniques are available to study neovascularization.
Collapse
Affiliation(s)
- Dimitry A Chistiakov
- a Department of Neurochemistry, Division of Basic and Applied Neurobiology , Serbsky Federal Medical Research Center of Psychiatry and Narcology , Moscow , Russia
| | - Alexandra A Melnichenko
- b Laboratory of Angiopathology , Institute of General Pathology and Pathophysiology, Russian Academy of Medical Sciences , Moscow , Russia
| | - Veronika A Myasoedova
- b Laboratory of Angiopathology , Institute of General Pathology and Pathophysiology, Russian Academy of Medical Sciences , Moscow , Russia
| | - Andrey V Grechko
- c Federal Research and Clinical Center of Intensive Care Medicine and Rehabilitology , Moscow , Russia
| | - Alexander N Orekhov
- b Laboratory of Angiopathology , Institute of General Pathology and Pathophysiology, Russian Academy of Medical Sciences , Moscow , Russia.,d Institute for Atherosclerosis Research, Skolkovo Innovative Center , Moscow , Russia
| |
Collapse
|
14
|
Adamus G. Can innate and autoimmune reactivity forecast early and advance stages of age-related macular degeneration? Autoimmun Rev 2017; 16:231-236. [PMID: 28137479 PMCID: PMC5334174 DOI: 10.1016/j.autrev.2017.01.005] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2016] [Accepted: 11/03/2016] [Indexed: 01/17/2023]
Abstract
Age-related macular degeneration (AMD) is a major cause of central vision loss in persons over 55years of age in developed countries. AMD is a complex disease in which genetic, environmental and inflammatory factors influence its onset and progression. Elevation in serum anti-retinal autoantibodies, plasma and local activation of complement proteins of the alternative pathway, and increase in secretion of proinflammatory cytokines have been seen over the course of disease. Genetic studies of AMD patients confirmed that genetic variants affecting the alternative complement pathway have a major influence on AMD risk. Because the heterogeneity of this disease, there is no sufficient strategy to identify the disease onset and progression sole based eye examination, thus identification of reliable serological biomarkers for diagnosis, prognosis and response to treatment by sampling patient's blood is necessary. This review provides an outline of the current knowledge on possible serological (autoantibodies, complement factors, cytokines, chemokines) and related genetic biomarkers relevant to the pathology of AMD, and discusses their application for prediction of disease activity and prognosis in AMD.
Collapse
Affiliation(s)
- Grazyna Adamus
- Ocular Immunology Laboratory, Casey Eye Institute, School of Medicine, Oregon Health and Science University, Portland, OR, USA.
| |
Collapse
|
15
|
Salomon RG. Carboxyethylpyrroles: From Hypothesis to the Discovery of Biologically Active Natural Products. Chem Res Toxicol 2016; 30:105-113. [PMID: 27750413 DOI: 10.1021/acs.chemrestox.6b00304] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Our research on the roles of lipid oxidation in human disease is guided by chemical intuition. For example, we postulated that 2-(ω-carboxyethyl)pyrrole (CEP) derivatives of primary amines would be produced through covalent adduction of a γ-hydroxyalkenal generated, in turn, through oxidative fragmentation of docosahexaenoates. Our studies confirmed the natural occurrence of this chemistry, and the biological activities of these natural products and their extensive involvements in human physiology (wound healing) and pathology (age-related macular degeneration, autism, atherosclerosis, sickle cell disease, and tumor growth) continue to emerge. This perspective recounts these discoveries and proposes new frontiers where further developments are likely. Perhaps more significantly, it depicts an effective chemistry-based approach to the discovery of novel biochemistry.
Collapse
Affiliation(s)
- Robert G Salomon
- Department of Chemistry, Case Western Reserve University , Cleveland, Ohio 44106, United States
| |
Collapse
|
16
|
Wang H, Linetsky M, Guo J, Yu AO, Salomon RG. Metabolism of 4-Hydroxy-7-oxo-5-heptenoic Acid (HOHA) Lactone by Retinal Pigmented Epithelial Cells. Chem Res Toxicol 2016; 29:1198-210. [PMID: 27355557 DOI: 10.1021/acs.chemrestox.6b00153] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
4-Hydroxy-7-oxo-5-heptenic acid (HOHA)-lactone is a biologically active oxidative truncation product released (t1/2 = 30 min at 37 °C) by nonenzymatic transesterification/deacylation from docosahexaenoate lipids. We now report that HOHA-lactone readily diffuses into retinal pigmented epithelial (RPE) cells where it is metabolized. A reduced glutathione (GSH) Michael adduct of HOHA-lactone is the most prominent metabolite detected by LC-MS in both the extracellular medium and cell lysates. This molecule appeared inside of ARPE-19 cells within seconds after exposure to HOHA-lactone. The intracellular level reached a maximum concentration at 30 min and then decreased with concomitant increases in its level in the extracellular medium, thus revealing a unidirectional export of the reduced GSH-HOHA-lactone adduct from the cytosol to extracellular medium. This metabolism is likely to modulate the involvement of HOHA-lactone in the pathogenesis of human diseases. HOHA-lactone is biologically active, e.g., low concentrations (0.1-1 μM) induce secretion of vascular endothelial growth factor (VEGF) from ARPE-19 cells. HOHA-lactone is also a precursor of 2-(ω-carboxyethyl)pyrrole (CEP) derivatives of primary amino groups in proteins and ethanolamine phospholipids that have significant pathological and physiological relevance to age-related macular degeneration (AMD), cancer, and wound healing. Both HOHA-lactone and the derived CEP can contribute to the angiogenesis that defines the neovascular "wet" form of AMD and that promotes the growth of tumors. While GSH depletion can increase the lethality of radiotherapy, because it will impair the metabolism of HOHA-lactone, the present study suggests that GSH depletion will also increase levels of HOHA-lactone and CEP that may promote recurrence of tumor growth.
Collapse
Affiliation(s)
- Hua Wang
- Department of Chemistry, Case Western Reserve University , Cleveland, Ohio 44106, United States
| | - Mikhail Linetsky
- Department of Chemistry, Case Western Reserve University , Cleveland, Ohio 44106, United States
| | - Junhong Guo
- Department of Chemistry, Case Western Reserve University , Cleveland, Ohio 44106, United States
| | - Annabelle O Yu
- Department of Chemistry, Case Western Reserve University , Cleveland, Ohio 44106, United States
| | - Robert G Salomon
- Department of Chemistry, Case Western Reserve University , Cleveland, Ohio 44106, United States
| |
Collapse
|
17
|
Abstract
Ageing, infections and inflammation result in oxidative stress that can irreversibly damage cellular structures. The oxidative damage of lipids in membranes or lipoproteins is one of these deleterious consequences that not only alters lipid function but also leads to the formation of neo-self epitopes - oxidation-specific epitopes (OSEs) - which are present on dying cells and damaged proteins. OSEs represent endogenous damage-associated molecular patterns that are recognized by pattern recognition receptors and the proteins of the innate immune system, and thereby enable the host to sense and remove dangerous biological waste and to maintain homeostasis. If this system is dysfunctional or overwhelmed, the accumulation of OSEs can trigger chronic inflammation and the development of diseases, such as atherosclerosis and age-related macular degeneration. Understanding the molecular components and mechanisms that are involved in this process will help to identify individuals with an increased risk of developing chronic inflammation, and will also help to indicate novel modes of therapeutic intervention.
Collapse
|
18
|
Guo J, Wang H, Hrinczenko B, Salomon RG. Efficient Quantitative Analysis of Carboxyalkylpyrrole Ethanolamine Phospholipids: Elevated Levels in Sickle Cell Disease Blood. Chem Res Toxicol 2016; 29:1187-97. [PMID: 27341308 DOI: 10.1021/acs.chemrestox.6b00152] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
γ-Hydroxy-α,β-unsaturated aldehydes, generated by oxidative damage of polyunsaturated phospholipids, form pyrrole derivatives that incorporate the ethanolamine phospholipid (EP) amino group such as 2-pentylpyrrole (PP)-EP and 2-(ω-carboxyalkyl)pyrrole (CAP)-EP derivatives: 2-(ω-carboxyethyl)pyrrole (CEP)-EP, 2-(ω-carboxypropyl)pyrrole (CPP)-EP, and 2-(ω-carboxyheptyl)pyrrole (CHP)-EP. Because EPs occur in vivo in various forms, a complex mixture of pyrrole-modified EPs with different molecular weights is expected to be generated. To provide a sensitive index of oxidative stress, all of the differences in mass related to the glycerophospholipid moieties were removed by releasing a single CAP-ethanolamine (ETN) or PP-ETN from each mixture by treatment with phospholipase D. Accurate quantization was achieved using the corresponding ethanolamine-d4 pyrroles as internal standards. The product mixture obtained by phospholipolysis of total blood phospholipids from sickle cell disease (SCD) patients was analyzed by LC-MS/MS. The method was applied to measure CAP-EP and PP-EP levels in blood plasma from clinical monitoring of SCD patients. We found uniformly elevated blood levels of CEP-EP (63.9 ± 9.7 nM) similar to mean levels in blood from age-related macular degeneration (AMD) patients (56.3 ± 37.1 nM), and 2-fold lower levels (27.6 ± 3.6 nM, n = 5) were detected in plasma from SCD patients hospitalized to treat a sickle cell crisis, although mean levels remain higher than those (12.1 ± 10.5 nM) detected in blood from healthy controls. Plasma levels of CPP-EPs from SCD clinic patients were 4-fold higher than those of SCD patients hospitalized to treat a sickle cell crisis (45.1 ± 10.9 nM, n = 5 versus 10.9 ± 3.4 nM, n = 6; p < 0.002). PP-EP concentration in plasma from SCD clinic patients is nearly 4.8-fold higher than its level in plasma samples from SCD patients hospitalized to treat a sickle cell crisis (7.06 ± 4.05 vs 1.48 ± 0.92 nM; p < 0.05). Because CAP-EPs promote angiogenesis and platelet activation, the elevated levels present in SCD blood can contribute to the hypercoaguability and vaso-occlusive events that are critical pathophysiologic features of SCD.
Collapse
Affiliation(s)
- Junhong Guo
- Department of Chemistry, Case Western Reserve University , Cleveland, Ohio 44106, United States
| | - Hua Wang
- Department of Chemistry, Case Western Reserve University , Cleveland, Ohio 44106, United States
| | - Borys Hrinczenko
- Division of Hematology and Oncology, Michigan State University , East Lansing, Michigan 48824, United States
| | - Robert G Salomon
- Department of Chemistry, Case Western Reserve University , Cleveland, Ohio 44106, United States
| |
Collapse
|
19
|
Du H, Xiao X, Stiles T, Douglas C, Ho D, Shaw PX. Novel Mechanistic Interplay between Products of Oxidative Stress and Components of the Complement System in AMD Pathogenesis. ACTA ACUST UNITED AC 2016; 6:43-50. [PMID: 27668132 PMCID: PMC5035113 DOI: 10.4236/ojoph.2016.61006] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/03/2022]
Abstract
Age-related macular degeneration (AMD) is a leading cause of vision loss affecting tens of millions of elderly worldwide. Early AMD includes soft drusen and pigmentary changes in the retinal pigment epithelium (RPE). As people age, such soft confluent drusen can progress into two forms of advanced AMD, geographic atrophy (GA, or dry AMD) or choroidal neovascularization (CNV, or wet AMD) and result in the loss of central vision. The exact mechanism for developing early AMD and progressing to advanced stage of disease is still largely unknown. However, significant evidence exists demonstrating a complex interplay of genetic and environmental factors as the cause of AMD progression. Together, complement factor H (CFH) and HTRA1/ARMS polymorphisms contribute to more than 50% of the genetic risk for AMD. Environmentally, oxidative stress from activities such as smoking has also demonstrated a powerful contribution to AMD progression. To extend our previous finding that genetic polymorphisms in CFH results in OxPLs and the risk-form of CFH (CFH Y402H) has reduced affinity for oxidized phospholipids, and subsequent diminished capacity which subsequently diminishes the capability to attenuate the inflammatory effects of these molecules, we compared the binding properties of CFH and CFH related protein 1 (CFHR1), which is also associated with disease risk, to OxPLs and their effects on modulating inflammation and lipids uptake. As both CFH-402H and CFHR1 are associated with increased risk to AMD, we hypothesized that like CFH-402H, CFHR1 contribution to AMD risk may also be due to its diminished affinity for OxPLs. Interestingly, we found that association of CFHR1 with OxPLs was not statistically different than CFH. However, binding of CFHR1 did not elicit the same protective benefits as CFH in that both inflammation and lipid uptake are unaffected by CFHR1 association with OxPLs. These findings demonstrate a novel and interesting complexity to the potential interplay between the complement system and oxidative stress byproducts, such as OxPLs, in the mechanistic contribution to AMD. Future work will aim to identify the molecular distinctions between CFH and CFHR1 which confer protection by the former, but not latter molecules. Understanding the molecular domains necessary for protection could provide interventional insights in the generation of novel therapeutics for AMD and other diseases associated with oxidative stress.
Collapse
Affiliation(s)
- Hongjun Du
- Department of Ophthalmology, Xijing Hospital, Xi'an, China; Department of Ophthalmology and Shiley Eye Institute, University of California San Diego, San Diego, USA
| | - Xu Xiao
- Sichuan Provincial People's Hospital, Chengdu, China
| | - Travis Stiles
- Department of Ophthalmology and Shiley Eye Institute, University of California San Diego, San Diego, USA
| | - Christopher Douglas
- Department of Ophthalmology and Shiley Eye Institute, University of California San Diego, San Diego, USA
| | - Daisy Ho
- Department of Ophthalmology and Shiley Eye Institute, University of California San Diego, San Diego, USA
| | - Peter X Shaw
- Department of Ophthalmology and Shiley Eye Institute, University of California San Diego, San Diego, USA
| |
Collapse
|
20
|
Xu J, Lu X, Shi GP. Vasa vasorum in atherosclerosis and clinical significance. Int J Mol Sci 2015; 16:11574-608. [PMID: 26006236 PMCID: PMC4463718 DOI: 10.3390/ijms160511574] [Citation(s) in RCA: 89] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2015] [Accepted: 05/11/2015] [Indexed: 12/12/2022] Open
Abstract
Atherosclerosis is a chronic inflammatory disease that leads to several acute cardiovascular complications with poor prognosis. For decades, the role of the adventitial vasa vasorum (VV) in the initiation and progression of atherosclerosis has received broad attention. The presence of VV neovascularization precedes the apparent symptoms of clinical atherosclerosis. VV also mediates inflammatory cell infiltration, intimal thickening, intraplaque hemorrhage, and subsequent atherothrombosis that results in stroke or myocardial infarction. Intraplaque neovessels originating from VV can be immature and hence susceptible to leakage, and are thus regarded as the leading cause of intraplaque hemorrhage. Evidence supports VV as a new surrogate target of atherosclerosis evaluation and treatment. This review provides an overview into the relationship between VV and atherosclerosis, including the anatomy and function of VV, the stimuli of VV neovascularization, and the available underlying mechanisms that lead to poor prognosis. We also summarize translational researches on VV imaging modalities and potential therapies that target VV neovascularization or its stimuli.
Collapse
Affiliation(s)
- Junyan Xu
- Second Clinical Medical College, Zhujiang Hospital and Southern Medical University, Guangzhou 510280, China.
| | - Xiaotong Lu
- Second Clinical Medical College, Zhujiang Hospital and Southern Medical University, Guangzhou 510280, China.
| | - Guo-Ping Shi
- Second Clinical Medical College, Zhujiang Hospital and Southern Medical University, Guangzhou 510280, China.
- Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
21
|
Kim YW, Yakubenko VP, West XZ, Gugiu GB, Renganathan K, Biswas S, Gao D, Crabb JW, Salomon RG, Podrez EA, Byzova TV. Receptor-Mediated Mechanism Controlling Tissue Levels of Bioactive Lipid Oxidation Products. Circ Res 2015; 117:321-32. [PMID: 25966710 DOI: 10.1161/circresaha.117.305925] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2014] [Accepted: 05/12/2015] [Indexed: 01/06/2023]
Abstract
RATIONALE Oxidative stress is an important contributing factor in several human pathologies ranging from atherosclerosis to cancer progression; however, the mechanisms underlying tissue protection from oxidation products are poorly understood. Oxidation of membrane phospholipids, containing the polyunsaturated fatty acid docosahexaenoic acid, results in the accumulation of an end product, 2-(ω-carboxyethyl)pyrrole (CEP), which was shown to have proangiogenic and proinflammatory functions. Although CEP is continuously accumulated during chronic processes, such as tumor progression and atherosclerosis, its level during wound healing return to normal when the wound is healed, suggesting the existence of a specific clearance mechanism. OBJECTIVE To identify the cellular and molecular mechanism for CEP clearance. METHODS AND RESULTS Here, we show that macrophages are able to bind, scavenge, and metabolize carboxyethylpyrrole derivatives of proteins but not structurally similar ethylpyrrole derivatives, demonstrating the high specificity of the process. F4/80(hi) and M2-skewed macrophages are much more efficient at CEP binding and scavenging compared with F4/80(lo) and M1-skewed macrophages. Depletion of macrophages leads to increased CEP accumulation in vivo. CEP binding and clearance are dependent on 2 receptors expressed by macrophages, CD36 and toll-like receptor 2. Although knockout of each individual receptor results in diminished CEP clearance, the lack of both receptors almost completely abrogates macrophages' ability to scavenge CEP derivatives of proteins. CONCLUSIONS Our study demonstrates the mechanisms of recognition, scavenging, and clearance of pathophysiologically active products of lipid oxidation in vivo, thereby contributing to tissue protection against products of oxidative stress.
Collapse
Affiliation(s)
- Young-Woong Kim
- From the Department of Molecular Cardiology, Lerner Research Institute (Y.-W.K., V.P.Y., X.Z.W., S.B., D.G., E.A.P., T.V.B.) and Department of Ophthalmology, Cole Eye Institute (R.K., J.W.C.), Cleveland Clinic, OH; and Department of Chemistry, Case Western Reserve University, Cleveland, OH (G.B.G., R.K., R.G.S.)
| | - Valentin P Yakubenko
- From the Department of Molecular Cardiology, Lerner Research Institute (Y.-W.K., V.P.Y., X.Z.W., S.B., D.G., E.A.P., T.V.B.) and Department of Ophthalmology, Cole Eye Institute (R.K., J.W.C.), Cleveland Clinic, OH; and Department of Chemistry, Case Western Reserve University, Cleveland, OH (G.B.G., R.K., R.G.S.)
| | - Xiaoxia Z West
- From the Department of Molecular Cardiology, Lerner Research Institute (Y.-W.K., V.P.Y., X.Z.W., S.B., D.G., E.A.P., T.V.B.) and Department of Ophthalmology, Cole Eye Institute (R.K., J.W.C.), Cleveland Clinic, OH; and Department of Chemistry, Case Western Reserve University, Cleveland, OH (G.B.G., R.K., R.G.S.)
| | - Gabriel B Gugiu
- From the Department of Molecular Cardiology, Lerner Research Institute (Y.-W.K., V.P.Y., X.Z.W., S.B., D.G., E.A.P., T.V.B.) and Department of Ophthalmology, Cole Eye Institute (R.K., J.W.C.), Cleveland Clinic, OH; and Department of Chemistry, Case Western Reserve University, Cleveland, OH (G.B.G., R.K., R.G.S.)
| | - Kutralanathan Renganathan
- From the Department of Molecular Cardiology, Lerner Research Institute (Y.-W.K., V.P.Y., X.Z.W., S.B., D.G., E.A.P., T.V.B.) and Department of Ophthalmology, Cole Eye Institute (R.K., J.W.C.), Cleveland Clinic, OH; and Department of Chemistry, Case Western Reserve University, Cleveland, OH (G.B.G., R.K., R.G.S.)
| | - Sudipta Biswas
- From the Department of Molecular Cardiology, Lerner Research Institute (Y.-W.K., V.P.Y., X.Z.W., S.B., D.G., E.A.P., T.V.B.) and Department of Ophthalmology, Cole Eye Institute (R.K., J.W.C.), Cleveland Clinic, OH; and Department of Chemistry, Case Western Reserve University, Cleveland, OH (G.B.G., R.K., R.G.S.)
| | - Detao Gao
- From the Department of Molecular Cardiology, Lerner Research Institute (Y.-W.K., V.P.Y., X.Z.W., S.B., D.G., E.A.P., T.V.B.) and Department of Ophthalmology, Cole Eye Institute (R.K., J.W.C.), Cleveland Clinic, OH; and Department of Chemistry, Case Western Reserve University, Cleveland, OH (G.B.G., R.K., R.G.S.)
| | - John W Crabb
- From the Department of Molecular Cardiology, Lerner Research Institute (Y.-W.K., V.P.Y., X.Z.W., S.B., D.G., E.A.P., T.V.B.) and Department of Ophthalmology, Cole Eye Institute (R.K., J.W.C.), Cleveland Clinic, OH; and Department of Chemistry, Case Western Reserve University, Cleveland, OH (G.B.G., R.K., R.G.S.)
| | - Robert G Salomon
- From the Department of Molecular Cardiology, Lerner Research Institute (Y.-W.K., V.P.Y., X.Z.W., S.B., D.G., E.A.P., T.V.B.) and Department of Ophthalmology, Cole Eye Institute (R.K., J.W.C.), Cleveland Clinic, OH; and Department of Chemistry, Case Western Reserve University, Cleveland, OH (G.B.G., R.K., R.G.S.)
| | - Eugene A Podrez
- From the Department of Molecular Cardiology, Lerner Research Institute (Y.-W.K., V.P.Y., X.Z.W., S.B., D.G., E.A.P., T.V.B.) and Department of Ophthalmology, Cole Eye Institute (R.K., J.W.C.), Cleveland Clinic, OH; and Department of Chemistry, Case Western Reserve University, Cleveland, OH (G.B.G., R.K., R.G.S.)
| | - Tatiana V Byzova
- From the Department of Molecular Cardiology, Lerner Research Institute (Y.-W.K., V.P.Y., X.Z.W., S.B., D.G., E.A.P., T.V.B.) and Department of Ophthalmology, Cole Eye Institute (R.K., J.W.C.), Cleveland Clinic, OH; and Department of Chemistry, Case Western Reserve University, Cleveland, OH (G.B.G., R.K., R.G.S.).
| |
Collapse
|
22
|
Wang H, Linetsky M, Guo J, Choi J, Hong L, Chamberlain AS, Howell SJ, Howes AM, Salomon RG. 4-Hydroxy-7-oxo-5-heptenoic Acid (HOHA) Lactone is a Biologically Active Precursor for the Generation of 2-(ω-Carboxyethyl)pyrrole (CEP) Derivatives of Proteins and Ethanolamine Phospholipids. Chem Res Toxicol 2015; 28:967-77. [PMID: 25793308 DOI: 10.1021/acs.chemrestox.5b00001] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
2-(ω-Carboxyethyl)pyrrole (CEP) derivatives of proteins were previously shown to have significant pathological and physiological relevance to age-related macular degeneration, cancer and wound healing. Previously, we showed that CEPs are generated in the reaction of ε-amino groups of protein lysyl residues with 1-palmityl-2-(4-hydroxy-7-oxo-5-heptenoyl)-sn-glycero-3-phosphatidylcholine (HOHA-PC), a lipid oxidation product uniquely generated by oxidative truncation of docosahexanenate-containing phosphatidylcholine. More recently, we found that HOHA-PC rapidly releases HOHA-lactone and 2-lyso-PC (t1/2 = 30 min at 37 °C) by nonenzymatic transesterification/deacylation. Now we report that HOHA-lactone reacts with Ac-Gly-Lys-OMe or human serum albumin to form CEP derivatives in vitro. Incubation of human red blood cell ghosts with HOHA-lactone generates CEP derivatives of membrane proteins and ethanolamine phospholipids. Quantitative analysis of the products generated in the reaction HOHA-PC with Ac-Gly-Lys-OMe showed that HOHA-PC mainly forms CEP-dipeptide that is not esterified to 2-lysophosphatidycholine. Thus, the HOHA-lactone pathway predominates over the direct reaction of HOHA-PC to produce the CEP-PC-dipeptide derivative. Myleoperoxidase/H2O2/NO2(-) promoted in vitro oxidation of either 1-palmityl-2-docosahexaneoyl-sn-glycero-3-phosphatidylcholine (DHA-PC) or docosahexaenoic acid (DHA) generates HOHA-lactone in yields of 0.45% and 0.78%, respectively. Lipid oxidation in human red blood cell ghosts also releases HOHA-lactone. Oxidative injury of ARPE-19 human retinal pigmented epithelial cells by exposure to H2O2 generated CEP derivatives. Treatment of ARPE-19 cells with HOHA-lactone generated CEP-modified proteins. Low (submicromolar), but not high, concentrations of HOHA-lactone promote increased vascular endothelial growth factor (VEGF) secretion by ARPE-19 cells. Therefore, HOHA-lactone not only serves as an intermediate for the generation of CEPs but also is a biologically active oxidative truncation product from docosahexaenoate lipids.
Collapse
Affiliation(s)
- Hua Wang
- Department of Chemistry, Case Western Reserve University, Cleveland, Ohio 44106, United States
| | - Mikhail Linetsky
- Department of Chemistry, Case Western Reserve University, Cleveland, Ohio 44106, United States
| | - Junhong Guo
- Department of Chemistry, Case Western Reserve University, Cleveland, Ohio 44106, United States
| | - Jaewoo Choi
- Department of Chemistry, Case Western Reserve University, Cleveland, Ohio 44106, United States
| | - Li Hong
- Department of Chemistry, Case Western Reserve University, Cleveland, Ohio 44106, United States
| | - Amanda S Chamberlain
- Department of Chemistry, Case Western Reserve University, Cleveland, Ohio 44106, United States
| | - Scott J Howell
- Department of Chemistry, Case Western Reserve University, Cleveland, Ohio 44106, United States
| | - Andrew M Howes
- Department of Chemistry, Case Western Reserve University, Cleveland, Ohio 44106, United States
| | - Robert G Salomon
- Department of Chemistry, Case Western Reserve University, Cleveland, Ohio 44106, United States
| |
Collapse
|
23
|
Comprehensive DNA adduct analysis reveals pulmonary inflammatory response contributes to genotoxic action of magnetite nanoparticles. Int J Mol Sci 2015; 16:3474-92. [PMID: 25658799 PMCID: PMC4346908 DOI: 10.3390/ijms16023474] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2014] [Revised: 01/06/2015] [Accepted: 01/30/2015] [Indexed: 12/17/2022] Open
Abstract
Nanosized-magnetite (MGT) is widely utilized in medicinal and industrial fields; however, its toxicological properties are not well documented. In our previous report, MGT showed genotoxicity in both in vitro and in vivo assay systems, and it was suggested that inflammatory responses exist behind the genotoxicity. To further clarify mechanisms underlying the genotoxicity, a comprehensive DNA adduct (DNA adductome) analysis was conducted using DNA samples derived from the lungs of mice exposed to MGT. In total, 30 and 42 types of DNA adducts were detected in the vehicle control and MGT-treated groups, respectively. Principal component analysis (PCA) against a subset of DNA adducts was applied and several adducts, which are deduced to be formed by inflammation or oxidative stress, as the case of etheno-deoxycytidine (εdC), revealed higher contributions to MGT exposure. By quantitative-LC-MS/MS analysis, εdC levels were significantly higher in MGT-treated mice than those of the vehicle control. Taken together with our previous data, it is suggested that inflammatory responses might be involved in the genotoxicity induced by MGT in the lungs of mice.
Collapse
|
24
|
Wang H, Guo J, West XZ, Bid HK, Lu L, Hong L, Jang GF, Zhang L, Crabb JW, Linetsky M, Salomon RG. Detection and biological activities of carboxyethylpyrrole ethanolamine phospholipids (CEP-EPs). Chem Res Toxicol 2014; 27:2015-22. [PMID: 25380349 PMCID: PMC4269404 DOI: 10.1021/tx500216a] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
![]()
Oxidation of docosahexaenoate phospholipids
produces 4-hydroxy-7-oxo-hept-5-eonyl
phospholipids (HOHA-PLs) that react with protein lysyl ε-amino
residues to generate 2-ω-carboxyethylpyrrole (CEP) derivatives,
endogenous factors that induce angiogenesis in the retina and tumors.
It seemed likely, but remained unproven, that HOHA-PLs react with
ethanolamine phospholipids (EPs) in vivo to generate
CEP-EPs. We now show that CEP-EPs are present in human blood at 4.6-fold
higher levels in age-related macular degeneration plasma than in normal
plasma. We also show that CEP-EPs are pro-angiogenic, inducing tube
formation by human umbilical vein endothelial cells by activating
Toll-like receptor 2. CEP-EP levels may be a useful biomarker for
clinical assessment of AMD risk and CEP-associated tumor progression
and a tool for monitoring the efficacy of therapeutic interventions.
Collapse
Affiliation(s)
- Hua Wang
- Department of Chemistry, Case Western Reserve University , Cleveland, Ohio 44106, United States
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Gounarides J, Cobb JS, Zhou J, Cook F, Yang X, Yin H, Meredith E, Rao C, Huang Q, Xu Y, Anderson K, De Erkenez A, Liao SM, Crowley M, Buchanan N, Poor S, Qiu Y, Fassbender E, Shen S, Woolfenden A, Jensen A, Cepeda R, Etemad-Gilbertson B, Giza S, Mogi M, Jaffee B, Azarian S. Lack of involvement of CEP adducts in TLR activation and in angiogenesis. PLoS One 2014; 9:e111472. [PMID: 25343517 PMCID: PMC4208838 DOI: 10.1371/journal.pone.0111472] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2013] [Accepted: 10/02/2014] [Indexed: 11/19/2022] Open
Abstract
Proteins that are post-translationally adducted with 2-(ω-carboxyethyl)pyrrole (CEP) have been proposed to play a pathogenic role in age-related macular degeneration, by inducing angiogenesis in a Toll Like Receptor 2 (TLR2)-dependent manner. We have investigated the involvement of CEP adducts in angiogenesis and TLR activation, to assess the therapeutic potential of inhibiting CEP adducts and TLR2 for ocular angiogenesis. As tool reagents, several CEP-adducted proteins and peptides were synthetically generated by published methodology and adduction was confirmed by NMR and LC-MS/MS analyses. Structural studies showed significant changes in secondary structure in CEP-adducted proteins but not the untreated proteins. Similar structural changes were also observed in the treated unadducted proteins, which were treated by the same adduction method except for one critical step required to form the CEP group. Thus some structural changes were unrelated to CEP groups and were artificially induced by the synthesis method. In biological studies, the CEP-adducted proteins and peptides failed to activate TLR2 in cell-based assays and in an in vivo TLR2-mediated retinal leukocyte infiltration model. Neither CEP adducts nor TLR agonists were able to induce angiogenesis in a tube formation assay. In vivo, treatment of animals with CEP-adducted protein had no effect on laser-induced choroidal neovascularization. Furthermore, in vivo inactivation of TLR2 by deficiency in Myeloid Differentiation factor 88 (Myd88) had no effect on abrasion-induced corneal neovascularization. Thus the CEP-TLR2 axis, which is implicated in other wound angiogenesis models, does not appear to play a pathological role in a corneal wound angiogenesis model. Collectively, our data do not support the mechanism of action of CEP adducts in TLR2-mediated angiogenesis proposed by others.
Collapse
Affiliation(s)
- John Gounarides
- Analytical Sciences, Novartis Institutes for Biomedical Research, Cambridge, MA, United States of America
| | - Jennifer S. Cobb
- Analytical Sciences, Novartis Institutes for Biomedical Research, Cambridge, MA, United States of America
| | - Jing Zhou
- Analytical Sciences, Novartis Institutes for Biomedical Research, Cambridge, MA, United States of America
| | - Frank Cook
- Analytical Sciences, Novartis Institutes for Biomedical Research, Cambridge, MA, United States of America
| | - Xuemei Yang
- Analytical Sciences, Novartis Institutes for Biomedical Research, Cambridge, MA, United States of America
| | - Hong Yin
- Analytical Sciences, Novartis Institutes for Biomedical Research, Cambridge, MA, United States of America
| | - Erik Meredith
- Global Discovery Chemistry, Novartis Institutes for Biomedical Research, Cambridge, MA, United States of America
| | - Chang Rao
- Analytical Sciences, Novartis Institutes for Biomedical Research, Cambridge, MA, United States of America
| | - Qian Huang
- Developmental and Metabolic Pathways, Novartis Institutes for Biomedical Research, Cambridge, MA, United States of America
| | - YongYao Xu
- Developmental and Metabolic Pathways, Novartis Institutes for Biomedical Research, Cambridge, MA, United States of America
| | - Karen Anderson
- Ophthalmology, Novartis Institutes for Biomedical Research, Cambridge, MA, United States of America
| | - Andrea De Erkenez
- Ophthalmology, Novartis Institutes for Biomedical Research, Cambridge, MA, United States of America
| | - Sha-Mei Liao
- Ophthalmology, Novartis Institutes for Biomedical Research, Cambridge, MA, United States of America
| | - Maura Crowley
- Ophthalmology, Novartis Institutes for Biomedical Research, Cambridge, MA, United States of America
| | - Natasha Buchanan
- Ophthalmology, Novartis Institutes for Biomedical Research, Cambridge, MA, United States of America
| | - Stephen Poor
- Ophthalmology, Novartis Institutes for Biomedical Research, Cambridge, MA, United States of America
| | - Yubin Qiu
- Ophthalmology, Novartis Institutes for Biomedical Research, Cambridge, MA, United States of America
| | - Elizabeth Fassbender
- Ophthalmology, Novartis Institutes for Biomedical Research, Cambridge, MA, United States of America
| | - Siyuan Shen
- Ophthalmology, Novartis Institutes for Biomedical Research, Cambridge, MA, United States of America
| | - Amber Woolfenden
- Ophthalmology, Novartis Institutes for Biomedical Research, Cambridge, MA, United States of America
| | - Amy Jensen
- Ophthalmology, Novartis Institutes for Biomedical Research, Cambridge, MA, United States of America
| | - Rosemarie Cepeda
- Ophthalmology, Novartis Institutes for Biomedical Research, Cambridge, MA, United States of America
| | - Bijan Etemad-Gilbertson
- Ophthalmology, Novartis Institutes for Biomedical Research, Cambridge, MA, United States of America
| | - Shelby Giza
- Ophthalmology, Novartis Institutes for Biomedical Research, Cambridge, MA, United States of America
| | - Muneto Mogi
- Global Discovery Chemistry, Novartis Institutes for Biomedical Research, Cambridge, MA, United States of America
| | - Bruce Jaffee
- Ophthalmology, Novartis Institutes for Biomedical Research, Cambridge, MA, United States of America
| | - Sassan Azarian
- Ophthalmology, Novartis Institutes for Biomedical Research, Cambridge, MA, United States of America
- * E-mail:
| |
Collapse
|
26
|
Lyzogubov VV, Bora NS, Tytarenko RG, Bora PS. Polyethylene glycol induced mouse model of retinal degeneration. Exp Eye Res 2014; 127:143-52. [PMID: 25088354 DOI: 10.1016/j.exer.2014.07.021] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2014] [Revised: 07/15/2014] [Accepted: 07/23/2014] [Indexed: 12/31/2022]
Abstract
Age-related macular degeneration (AMD) is a leading cause of irreversible blindness. This study was done to characterize dry AMD-like changes in mouse retinal pigment epithelium (RPE) and retina after polyethylene glycol (PEG) treatment. We injected male C57BL/6 mice subretinally with PBS, 0.025, 0.25, 0.5 and 1.0 mg of PEG-400 and the animals were sacrificed on day 5. Eyes were harvested and processed for histological analysis. In all other experiments 0.5 mg PEG was injected and animals were sacrificed on days 1, 3, 5 or 14. Paraffin, 5 μm and plastic, 1 μm and 80 nm sections were used for further analysis. Subretinal injection of 0.5 mg PEG induced a 32% reduction of outer nuclear layer (ONL) thickness, 61% decrease of photoreceptor outer and inner segment length, 49% decrease of nuclear density in the ONL and 31% increase of RPE cell density by day 5 after injection. The maximum level of TUNEL positive nuclei in the ONL (6.8 + 1.99%) was detected at day 5 after PEG injection and co-localized with Casp3act. Histological signs of apoptosis were observed in the ONL by light or electron microscopy. Degeneration of RPE cells was found in PEG injected eyes. Gene expression data identified several genes reported to be involved in human AMD. C3, Cfi, Serping1, Mmp9, Htra1 and Lpl were up-regulated in PEG injected eyes compared to PBS controls. PEG leads to morphological and gene expression changes in RPE and retina consistent with dry AMD. This model will be useful to investigate dry AMD pathogenesis and treatment.
Collapse
Affiliation(s)
- Valeriy V Lyzogubov
- Department of Ophthalmology, Jones Eye Institute, Pat & Willard Walker Eye Research Center, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Nalini S Bora
- Department of Ophthalmology, Jones Eye Institute, Pat & Willard Walker Eye Research Center, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Ruslana G Tytarenko
- Department of Ophthalmology, Jones Eye Institute, Pat & Willard Walker Eye Research Center, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Puran S Bora
- Department of Ophthalmology, Jones Eye Institute, Pat & Willard Walker Eye Research Center, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA.
| |
Collapse
|
27
|
Mullins RF, Schoo DP, Sohn EH, Flamme-Wiese MJ, Workamelahu G, Johnston RM, Wang K, Tucker BA, Stone EM. The membrane attack complex in aging human choriocapillaris: relationship to macular degeneration and choroidal thinning. THE AMERICAN JOURNAL OF PATHOLOGY 2014; 184:3142-53. [PMID: 25204844 DOI: 10.1016/j.ajpath.2014.07.017] [Citation(s) in RCA: 165] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/18/2014] [Revised: 06/18/2014] [Accepted: 07/10/2014] [Indexed: 01/19/2023]
Abstract
Age-related macular degeneration (AMD) is a common disease that can result in severe visual impairment. Abnormal regulation of the complement system has been implicated in its pathogenesis, and CFH polymorphisms contribute substantially to risk. How these polymorphisms exert their effects is poorly understood. We performed enzyme-linked immunosorbent assay (ELISA) analysis on young, aged, and AMD choroids to determine the abundance of the membrane attack complex (MAC) and performed immunofluorescence studies on eyes from 117 donors to evaluate the MAC in aging, early AMD, and advanced AMD. Morphometric studies were performed on eyes with high- or low-risk CFH genotypes. ELISA confirmed that MAC increases significantly with aging and with AMD. MAC was localized to Bruch's membrane and the choriocapillaris and was detectable at low levels as early as 5 years of age. Hard drusen were labeled with anti-MAC antibody, but large or confluent drusen and basal deposits were generally unlabeled. Labeling of retinal pigment epithelium was observed in some cases of advanced AMD, but not in early disease. Eyes homozygous for the high-risk CFH genotype had thinner choroids than low-risk homozygotes (P < 0.05). These findings suggest that increased complement activation in AMD and in high-risk genotypes can lead to loss of endothelial cells in early AMD. Treatments to protect the choriocapillaris in early AMD are needed.
Collapse
Affiliation(s)
- Robert F Mullins
- Department of Ophthalmology and Visual Sciences, The University of Iowa, Iowa City, Iowa; Stephen A. Wynn Institute for Vision Research, The University of Iowa, Iowa City, Iowa.
| | - Desi P Schoo
- Department of Ophthalmology and Visual Sciences, The University of Iowa, Iowa City, Iowa; Stephen A. Wynn Institute for Vision Research, The University of Iowa, Iowa City, Iowa
| | - Elliott H Sohn
- Department of Ophthalmology and Visual Sciences, The University of Iowa, Iowa City, Iowa; Stephen A. Wynn Institute for Vision Research, The University of Iowa, Iowa City, Iowa
| | - Miles J Flamme-Wiese
- Department of Ophthalmology and Visual Sciences, The University of Iowa, Iowa City, Iowa; Stephen A. Wynn Institute for Vision Research, The University of Iowa, Iowa City, Iowa
| | - Grefachew Workamelahu
- Department of Ophthalmology and Visual Sciences, The University of Iowa, Iowa City, Iowa; Stephen A. Wynn Institute for Vision Research, The University of Iowa, Iowa City, Iowa
| | - Rebecca M Johnston
- Department of Ophthalmology and Visual Sciences, The University of Iowa, Iowa City, Iowa; Stephen A. Wynn Institute for Vision Research, The University of Iowa, Iowa City, Iowa
| | - Kai Wang
- Stephen A. Wynn Institute for Vision Research, The University of Iowa, Iowa City, Iowa; Department of Biostatistics, The University of Iowa, Iowa City, Iowa
| | - Budd A Tucker
- Department of Ophthalmology and Visual Sciences, The University of Iowa, Iowa City, Iowa; Stephen A. Wynn Institute for Vision Research, The University of Iowa, Iowa City, Iowa
| | - Edwin M Stone
- Department of Ophthalmology and Visual Sciences, The University of Iowa, Iowa City, Iowa; Stephen A. Wynn Institute for Vision Research, The University of Iowa, Iowa City, Iowa
| |
Collapse
|
28
|
Saeed AM, Duffort S, Ivanov D, Wang H, Laird JM, Salomon RG, Cruz-Guilloty F, Perez VL. The oxidative stress product carboxyethylpyrrole potentiates TLR2/TLR1 inflammatory signaling in macrophages. PLoS One 2014; 9:e106421. [PMID: 25184331 PMCID: PMC4153630 DOI: 10.1371/journal.pone.0106421] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2014] [Accepted: 07/29/2014] [Indexed: 12/18/2022] Open
Abstract
Oxidative stress is key in the pathogenesis of several diseases including age-related macular degeneration (AMD), atherosclerosis, diabetes, and Alzheimer's disease. It has previously been established that a lipid peroxidation product, carboxyethylpyrrole (CEP), accumulates in the retinas of AMD patients. Retinal infiltrating macrophages also accumulate in the retinas of both AMD patients and in a murine model of AMD. We therefore investigated the ability of CEP-adducts to activate innate immune signaling in murine bone-marrow derived macrophages (BMDMs). We found that CEP specifically synergizes with low-dose TLR2-agonists (but not agonists for other TLRs) to induce production of inflammatory cytokines. Moreover, CEP selectively augments TLR2/TLR1-signaling instead of TLR2/TLR6-signaling. These studies uncover a novel synergistic inflammatory relationship between an endogenously produced oxidation molecule and a pathogen-derived product, which may have implications in the AMD disease process and other oxidative stress-driven pathologies.
Collapse
Affiliation(s)
- Ali M. Saeed
- Sheila and David Fuente Program in Cancer Biology, University of Miami Miller School of Medicine, Miami, Florida, United States of America
| | - Stephanie Duffort
- Bascom Palmer Eye Institute, Department of Ophthalmology, University of Miami Miller School of Medicine, Miami, Florida, United States of America
| | - Dmitry Ivanov
- Bascom Palmer Eye Institute, Department of Ophthalmology, University of Miami Miller School of Medicine, Miami, Florida, United States of America
| | - Hua Wang
- Department of Chemistry, Case Western Reserve University, Cleveland, Ohio, United States of America
| | - James M. Laird
- Department of Chemistry, Case Western Reserve University, Cleveland, Ohio, United States of America
| | - Robert G. Salomon
- Department of Chemistry, Case Western Reserve University, Cleveland, Ohio, United States of America
| | - Fernando Cruz-Guilloty
- Bascom Palmer Eye Institute, Department of Ophthalmology, University of Miami Miller School of Medicine, Miami, Florida, United States of America
- Department of Microbiology and Immunology, University of Miami Miller School of Medicine, Miami, Florida, United States of America
- * E-mail: (VLP); (FC-G)
| | - Victor L. Perez
- Bascom Palmer Eye Institute, Department of Ophthalmology, University of Miami Miller School of Medicine, Miami, Florida, United States of America
- Department of Microbiology and Immunology, University of Miami Miller School of Medicine, Miami, Florida, United States of America
- * E-mail: (VLP); (FC-G)
| |
Collapse
|
29
|
Nowak JZ. Oxidative stress, polyunsaturated fatty acids-derived oxidation products and bisretinoids as potential inducers of CNS diseases: focus on age-related macular degeneration. Pharmacol Rep 2014; 65:288-304. [PMID: 23744414 DOI: 10.1016/s1734-1140(13)71005-3] [Citation(s) in RCA: 90] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2012] [Revised: 01/31/2013] [Indexed: 12/12/2022]
Abstract
Many pathologies of the central nervous system (CNS) originate from excess of reactive free radicals, notably reactive oxygen species (ROS), and oxidative stress. A phenomenon which usually runs in parallel with oxidative stress is unsaturated lipid peroxidation, which, via a chain reaction, contributes to the progression of disbalanced redox homeostasis. Among long-chain (LC) polyunsaturated fatty acids (PUFAs) abundantly occurring in the CNS, docosahexaenoic acid (DHA), a member of ω-3 LC-PUFAs, deserves special attention, as it is avidly retained and uniquely concentrated in the nervous system, particularly in retinal photoreceptors and synaptic membranes; owing to the presence of the six double bonds between carbon atoms in its polyene chain (C=C), DHA is exquisitely sensitive to oxidative damage. In addition to oxidative stress and LC-PUFAs peroxidation, other stress-related mechanisms may also contribute to the development of various CNS malfunctions, and a good example of such mechanisms is the process of lipofuscin formation occurring particularly in the retina, an integral part of the CNS. The retinal lipofuscin is formed and accumulated by the retinal pigment epithelial (RPE) cells as a consequence of both visual process taking place in photoreceptor-RPE functional complex and metabolic insufficiency of RPE lysosomal compartment. Among various retinal lipofuscin constituents, bisretinoids, originating from all-trans retinal substrate--a photometabolite of visual pigment cofactor 11-cis-retinal (responsible for photon capturing), are endowed with cytotoxic and complement-activating potential which increases upon illumination and oxidation. This survey deals with oxidative stress, PUFAs (especially DHA) peroxidation products of carboxyalkylpyrrole type and bisretinoids as potential inducers of the CNS pathology. A focus is put on vision-threatening disease, i.e., age-related macular degeneration (AMD), as an example of the CNS disorder whose pathogenesis has strong background in both oxidative stress and lipid peroxidation products.
Collapse
Affiliation(s)
- Jerzy Z Nowak
- Institute of Pharmacology, Polish Academy of Sciences, Scientific Board, Smętna 12, PL 31-343 Kraków, Poland.
| |
Collapse
|
30
|
Abstract
Despite the damaging effect on tissues at a high concentration, it has been gradually established that oxidative stress plays a positive role during angiogenesis. In adults, physiological or pathological angiogenesis is initiated by tissue demands for oxygen and nutrients, resulting in a hypoxia/reoxygenation cycle, which, in turn promotes the formation of reactive oxygen species (ROS). The ROS can be generated either endogenously, through mitochondrial electron transport chain reactions and nicotinamide adenine dinucleotide phosphate oxidase, or exogenously, resulting from exposure to environmental agents, such as ultraviolet or ionizing radiation. In many conditions, ROS promotes angiogenesis, either directly or via the generation of active oxidation products, including peroxidized lipids. The latter lipid metabolites are generated in excess during atherosclerosis, thereby linking atherogenic processes and pathological angiogenesis. Although the main mechanism of oxidative stress-induced angiogenesis involves hypoxia-inducible factor/vascular endothelial growth factor (VEGF) signaling, recent studies have identified several pathways that are VEGF-independent. This review aims to provide a summary of the past and present views on the role of oxidative stress as a mediator and modulator of angiogenesis, and to highlight newly identified mechanisms.
Collapse
|
31
|
Pizzimenti S, Ciamporcero E, Daga M, Pettazzoni P, Arcaro A, Cetrangolo G, Minelli R, Dianzani C, Lepore A, Gentile F, Barrera G. Interaction of aldehydes derived from lipid peroxidation and membrane proteins. Front Physiol 2013; 4:242. [PMID: 24027536 PMCID: PMC3761222 DOI: 10.3389/fphys.2013.00242] [Citation(s) in RCA: 246] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2013] [Accepted: 08/15/2013] [Indexed: 01/07/2023] Open
Abstract
A great variety of compounds are formed during lipid peroxidation of polyunsaturated fatty acids of membrane phospholipids. Among them, bioactive aldehydes, such as 4-hydroxyalkenals, malondialdehyde (MDA) and acrolein, have received particular attention since they have been considered as toxic messengers that can propagate and amplify oxidative injury. In the 4-hydroxyalkenal class, 4-hydroxy-2-nonenal (HNE) is the most intensively studied aldehyde, in relation not only to its toxic function, but also to its physiological role. Indeed, HNE can be found at low concentrations in human tissues and plasma and participates in the control of biological processes, such as signal transduction, cell proliferation, and differentiation. Moreover, at low doses, HNE exerts an anti-cancer effect, by inhibiting cell proliferation, angiogenesis, cell adhesion and by inducing differentiation and/or apoptosis in various tumor cell lines. It is very likely that a substantial fraction of the effects observed in cellular responses, induced by HNE and related aldehydes, be mediated by their interaction with proteins, resulting in the formation of covalent adducts or in the modulation of their expression and/or activity. In this review we focus on membrane proteins affected by lipid peroxidation-derived aldehydes, under physiological and pathological conditions.
Collapse
Affiliation(s)
- Stefania Pizzimenti
- Dipartimento di Scienze Cliniche e Biologiche, Università di Torino Torino, Italy
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Perez VL, Saeed AM, Tan Y, Urbieta M, Cruz-Guilloty F. The eye: A window to the soul of the immune system. J Autoimmun 2013; 45:7-14. [PMID: 23871641 DOI: 10.1016/j.jaut.2013.06.011] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2013] [Accepted: 06/18/2013] [Indexed: 01/17/2023]
Abstract
The eye is considered as an immune privileged site, and with good reason. It has evolved a variety of molecular and cellular mechanisms that limit immune responses to preserve vision. For example, the cornea is mainly protected from autoimmunity by the lack of blood and lymphatic vessels, whereas the retina-blood barrier is maintained in an immunosuppressive state by the retinal pigment epithelium. However, there are several scenarios in which immune privilege is altered and the eye becomes susceptible to immune attack. In this review, we highlight the role of the immune system in two clinical conditions that affect the anterior and posterior segments of the eye: corneal transplantation and age-related macular degeneration. Interestingly, crosstalk between the innate and adaptive immune systems is critical in both acute and chronic inflammatory responses in the eye, with T cells playing a central role in combination with neutrophils and macrophages. In addition, we emphasize the advantage of using the eye as a model for in vivo longitudinal imaging of the immune system in action. Through this technique, it has been possible to identify functionally distinct intra-graft motility patterns of responding T cells, as well as the importance of chemokine signaling in situ for T cell activation. The detailed study of ocular autoimmunity could provide novel therapeutic strategies for blinding diseases while also providing more general information on acute versus chronic inflammation.
Collapse
Affiliation(s)
- V L Perez
- Laboratory of Ocular Immunology and Transplantation, Bascom Palmer Eye Institute, University of Miami Miller School of Medicine, Miami, FL 33136, USA; Department of Microbiology & Immunology, University of Miami Miller School of Medicine, Miami, FL 33136, USA.
| | | | | | | | | |
Collapse
|
33
|
McDowell RE, McGeown JG, Stitt AW, Curtis TM. Therapeutic potential of targeting lipid aldehydes and lipoxidation end-products in the treatment of ocular disease. Future Med Chem 2013; 5:189-211. [PMID: 23360143 DOI: 10.4155/fmc.12.202] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Lipoxidation reactions and the subsequent accumulation of advanced lipoxidation end products (ALEs) have been implicated in the pathogenesis of many of the leading causes of visual impairment. Here, we begin by outlining some of the major lipid aldehydes produced through lipoxidation reactions, the ALEs formed upon their reaction with proteins, and the endogenous aldehyde metabolizing enzymes involved in protecting cells against lipoxidation mediated damage. Discussions are subsequently focused on the clinical and experimental evidence supporting the contribution of lipid aldehydes and ALEs in the development of ocular diseases. From these discussions, it is clear that inhibition of lipoxidation reactions and ALE formation could represent a new therapeutic avenue for the treatment of a broad range of ocular disorders. Current and emerging pharmacological strategies to prevent or neutralize the effects of lipid aldehydes and ALEs are therefore considered, with particular emphasis on the potential of these drugs for treatment of diseases of the eye.
Collapse
Affiliation(s)
- Rosemary E McDowell
- Centre for Vision & Vascular Science, Queen's University of Belfast, Institute of Clinical Sciences, The Royal Victoria Hospital, Grosvenor Road, Belfast BT12 6BA, Northern Ireland, UK
| | | | | | | |
Collapse
|
34
|
Abstract
Free radical-induced oxidation of membrane phospholipids generates complex mixtures of oxidized phospholipids (oxPLs). The combinatorial operation of a few dozen reaction types on a few dozen phospholipid structures results in the production of a dauntingly vast diversity of oxPL molecular species. Structural identification of the individual oxPL in these mixtures is a redoubtable challenge that is absolutely essential to allow determination of the biological activities of individual species. With an emphasis on cardiovascular consequences, this Review focuses on biological activities of oxPLs whose molecular structures are known and highlights 2 diametrically opposite approaches that were used to determine those structures, that is, (1) the classic approach from bioactivity of a complex mixture to isolation and structural characterization of the active molecule followed by confirmation of the structure by unambiguous chemical synthesis and (2) hypothesis of products that are likely to be generated by lipid oxidation, followed by synthesis, and then detection in vivo guided by the availability of authentic standards, and last, characterization of biological activities. Especially important for the application of the second paradigm is the capability of LC-MS/MS and derivatizations to selectively detect and quantify specific oxPL in complex mixtures, without the need for their isolation or complete separation. This technology can provide strong evidence for identity by comparisons with pure, well-characterized samples available by chemical syntheses. Those pure samples are critical for determining the biological activities attributable to specific molecular species of oxPLs in the complex mixtures generated in vivo as a consequence of oxidative stress.
Collapse
Affiliation(s)
- Robert G Salomon
- Department of Chemistry, Case Western Reserve University, Cleveland, OH 44106, USA.
| |
Collapse
|
35
|
Lee S, Birukov KG, Romanoski CE, Springstead JR, Lusis AJ, Berliner JA. Role of phospholipid oxidation products in atherosclerosis. Circ Res 2012; 111:778-99. [PMID: 22935534 DOI: 10.1161/circresaha.111.256859] [Citation(s) in RCA: 150] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
There is increasing clinical evidence that phospholipid oxidation products (Ox-PL) play a role in atherosclerosis. This review focuses on the mechanisms by which Ox-PL interact with endothelial cells, monocyte/macrophages, platelets, smooth muscle cells, and HDL to promote atherogenesis. In the past few years major progress has been made in identifying these mechanisms. It has been recognized that Ox-PL promote phenotypic changes in these cell types that have long-term consequences for the vessel wall. Individual Ox-PL responsible for specific cellular effects have been identified. A model of the configuration of bioactive truncated Ox-PL within membranes has been developed that demonstrates that the oxidized fatty acid moiety protrudes into the aqueous phase, rendering it accessible for receptor recognition. Receptors and signaling pathways for individual Ox-PL species are now determined and receptor independent signaling pathways identified. The effects of Ox-PL are mediated both by gene regulation and transcription independent processes. It has now become apparent that Ox-PL affects multiple genes and pathways, some of which are proatherogenic and some are protective. However, at concentrations that are likely present in the vessel wall in atherosclerotic lesions, the effects promote atherogenesis. There have also been new insights on enzymes that metabolize Ox-PL and the significance of these enzymes for atherosclerosis. With the knowledge we now have of the regulation and effects of Ox-PL in different vascular cell types, it should be possible to design experiments to test the role of specific Ox-PL on the development of atherosclerosis.
Collapse
Affiliation(s)
- Sangderk Lee
- Department of Pathology, University of California-Los Angeles, MRL 4760, 675 Charles E. Young Dr. S., Los Angeles, CA 90095, USA
| | | | | | | | | | | |
Collapse
|
36
|
DeVolder RJ, Zill AT, Jeong JH, Kong H. Microfabrication of proangiogenic cell-Laden alginate-g-Pyrrole hydrogels. Biomaterials 2012; 33:7718-26. [DOI: 10.1016/j.biomaterials.2012.07.001] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2012] [Accepted: 07/01/2012] [Indexed: 01/03/2023]
|
37
|
Ullery JC, Marnett LJ. Protein modification by oxidized phospholipids and hydrolytically released lipid electrophiles: Investigating cellular responses. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2012; 1818:2424-35. [PMID: 22562025 DOI: 10.1016/j.bbamem.2012.04.014] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/10/2012] [Revised: 04/05/2012] [Accepted: 04/17/2012] [Indexed: 12/17/2022]
Abstract
Oxygen is essential for the growth and function of mammalian cells. However, imbalances in oxygen or abnormalities in the ability of a cell to respond to oxygen levels can result in oxidative stress. Oxidative stress plays an important role in a number of diseases including atherosclerosis, rheumatoid arthritis, cancer, neurodegenerative diseases and asthma. When membrane lipids are exposed to high levels of oxygen or derived oxidants, they undergo lipid peroxidation to generate oxidized phospholipids (oxPL). Continual exposure to oxidants and decomposition of oxPL results in the formation of reactive electrophiles, such as 4-hydroxy-2-nonenal (HNE). Reactive lipid electrophiles have been shown to covalently modify DNA and proteins. Furthermore, exposure of cells to lipid electrophiles results in the activation of cytoprotective signaling pathways in order to promote cell survival and recovery from oxidant stress. However, if not properly managed by cellular detoxification mechanisms, the continual exposure of cells to electrophiles results in cytotoxicity. The following perspective will discuss the biological importance of lipid electrophile protein adducts including current strategies employed to identify and isolate protein adducts of lipid electrophiles as well as approaches to define cellular signaling mechanisms altered upon exposure to electrophiles. This article is part of a Special Issue entitled: Oxidized phospholipids-their properties and interactions with proteins.
Collapse
Affiliation(s)
- Jody C Ullery
- Department of Biochemistry, Vanderbilt Institute of Chemical Biology, Nashville, TN, USA
| | | |
Collapse
|
38
|
Salomon RG, Gu X. Critical insights into cardiovascular disease from basic research on the oxidation of phospholipids: the γ-hydroxyalkenal phospholipid hypothesis. Chem Res Toxicol 2011; 24:1791-802. [PMID: 21870852 DOI: 10.1021/tx200207z] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Basic research, exploring the hypothesis that γ-hydroxyalkenal phospholipids are generated in vivo through oxidative cleavage of polyunsaturated phospholipids, is delivering a bonanza of molecular mechanistic insights into cardiovascular disease. Rather than targeting a specific pathology, these studies were predicated on the presumption that a fundamental understanding of lipid oxidation is likely to provide critical insights into disease processes. This investigational approach, from the chemistry of biomolecules to disease phenotype, that complements the more common opposite paradigm, is proving remarkably productive.
Collapse
Affiliation(s)
- Robert G Salomon
- Department of Chemistry, Case Western Reserve University, Cleveland, Ohio 44106-7078, USA.
| | | |
Collapse
|