1
|
Feng C, Zhang L, Zhou X, Lu S, Guo R, Song C, Zhang X. Redox imbalance drives magnetic property and function changes in mice. Redox Biol 2025; 81:103561. [PMID: 40020452 PMCID: PMC11910372 DOI: 10.1016/j.redox.2025.103561] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2025] [Revised: 02/05/2025] [Accepted: 02/20/2025] [Indexed: 03/03/2025] Open
Abstract
The magnetic properties of substances directly determine their response to an externally applied magnetic field, which are closely associated with magnetoreception, magnetic resonance imaging (MRI), and magnetic bioeffects. However, people's understanding of the magnetic properties of living organisms remains limited. In this study, we utilized NRF2 (nuclear factor erythroid 2-related factor 2) deficient mice to investigate the contribution of redox (oxidation-reduction) homeostasis, in which the key process is the transfer of electron, a direct target of magnetic field and origin of paramagnetism. Our results show that the NRF2-/- mice exhibit significantly altered systemic redox state, accompanied by increased magnetic susceptibility, particularly in the liver and spleen. Further analyses reveal that the levels of paramagnetic reactive oxygen species (ROS) in these tissues are markedly elevated compared to wild-type mice. Moreover, the concentrations of Fe2+ and Fe3+ are significantly elevated in NRF2-/- mice, which are directly correlated with the increased magnetic susceptibility. The disrupted redox balance in NRF2-/- mice not only exacerbates oxidative stress and iron deposition, but also induces impairment to the liver and spleen. The findings highlight the combined effects of ROS and iron metabolism in driving magnetic susceptibility changes, providing valuable theoretical insights for further research into magnetic bioeffects and organ-specific sensitivity to magnetic fields.
Collapse
Affiliation(s)
- Chuanlin Feng
- High Magnetic Field Laboratory, CAS Key Laboratory of High Magnetic Field and Ion Beam Physical Biology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, 230031, China; Science Island Branch of Graduate School, University of Science and Technology of China, Hefei, 230026, China
| | - Lei Zhang
- High Magnetic Field Laboratory, CAS Key Laboratory of High Magnetic Field and Ion Beam Physical Biology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, 230031, China
| | - Xiaoyuan Zhou
- Institutes of Physical Science and Information Technology, Anhui University, Hefei, 230039, China
| | - Shiyu Lu
- High Magnetic Field Laboratory, CAS Key Laboratory of High Magnetic Field and Ion Beam Physical Biology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, 230031, China; Science Island Branch of Graduate School, University of Science and Technology of China, Hefei, 230026, China
| | - Ruowen Guo
- High Magnetic Field Laboratory, CAS Key Laboratory of High Magnetic Field and Ion Beam Physical Biology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, 230031, China; Science Island Branch of Graduate School, University of Science and Technology of China, Hefei, 230026, China
| | - Chao Song
- High Magnetic Field Laboratory, CAS Key Laboratory of High Magnetic Field and Ion Beam Physical Biology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, 230031, China.
| | - Xin Zhang
- High Magnetic Field Laboratory, CAS Key Laboratory of High Magnetic Field and Ion Beam Physical Biology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, 230031, China; Science Island Branch of Graduate School, University of Science and Technology of China, Hefei, 230026, China; Institutes of Physical Science and Information Technology, Anhui University, Hefei, 230039, China.
| |
Collapse
|
2
|
Haidery F, Lambertini L, Tse I, Dodda S, Garcia-Ocaña A, Scott DK, Baumel-Alterzon S. NRF2 deficiency leads to inadequate beta cell adaptation during pregnancy and gestational diabetes. Redox Biol 2025; 81:103566. [PMID: 40054060 PMCID: PMC11930207 DOI: 10.1016/j.redox.2025.103566] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2024] [Revised: 02/13/2025] [Accepted: 02/24/2025] [Indexed: 03/12/2025] Open
Abstract
The late stages of mammalian pregnancy are accompanied by a mild increase in insulin resistance likely due to enhanced glucose demand of the growing fetus. Therefore, as an adaptive process to maintain euglycemia during pregnancy, maternal β-cell mass expands leading to increased insulin release. Defects in functional β-cell adaptive expansion during pregnancy can lead to gestational diabetes mellitus (GDM). While the exact mechanisms that promote GDM are poorly understood, GDM is associated with inadequate functional β-cell mass expansion and with a systematic increase of oxidative stress. Here, we show that NRF2 levels are upregulated in mouse β-cells at gestational day 15 (GD15). Inducible β-cell-specific Nrf2 deleted (βNrf2KO) mice display reduced β-cell proliferation, increased β-cell oxidative stress and lipid peroxidation, compromised β-cell function, and elevated β-cell death, leading to impaired β-cell mass expansion and dysregulated glucose homeostasis towards the end of pregnancy. Importantly, the gestational hormone 17-β-estradiol (E2) increases NRF2 levels, and downregulation of NRF2 suppresses E2-induced protection of β-cells against oxidative stress, suggesting that E2 exerts its antioxidant effects through activation of NRF2 signaling in β-cells. Collectively, these data highlight the critical role of NRF2 in regulating oxidative stress during the adaptive response of β-cells in pregnancy and identify NRF2 as a potential therapeutic target for GDM treatment.
Collapse
Affiliation(s)
- Fatema Haidery
- Diabetes, Obesity and Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Luca Lambertini
- Diabetes, Obesity and Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Isabelle Tse
- Diabetes, Obesity and Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Sriya Dodda
- Diabetes, Obesity and Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Adolfo Garcia-Ocaña
- Department of Molecular and Cellular Endocrinology, Arthur Riggs Diabetes & Metabolism Research Institute at City of Hope, Duarte, CA, USA
| | - Donald K Scott
- Diabetes, Obesity and Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Sharon Baumel-Alterzon
- Diabetes, Obesity and Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Mindich Child Health and Development Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Department of Molecular and Cellular Endocrinology, Arthur Riggs Diabetes & Metabolism Research Institute at City of Hope, Duarte, CA, USA.
| |
Collapse
|
3
|
Okutan E, Güleç İ, Şengelen A, Karagöz-Güzey F, Eren B, Tufan A, Özcan TB, Önay-Uçar E. Aloperine treatment attenuates acute spinal cord injury by reducing oxidative, inflammatory, and apoptotic responses via PI3K/AKT/NF-κB signaling in a rat contusion model. Neurosci Lett 2025; 854:138203. [PMID: 40132779 DOI: 10.1016/j.neulet.2025.138203] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2025] [Revised: 03/09/2025] [Accepted: 03/16/2025] [Indexed: 03/27/2025]
Abstract
Spinal cord injury (SCI) is a severe condition that can result in nerve damage, impaired motor or sensory function, and ultimately a high mortality rate for injured individuals. High oxidative and inflammatory responses are closely linked to poor prognosis and can influence the recovery of neurological functions. Therefore, overcoming these processes early is a valuable therapy approach for SCI. Aloperine (ALO) is a quinolizidine-type alkaloid with numerous pharmacological activities, including antioxidant, anti-inflammatory, and neuroprotective effects. However, the role of ALO in SCI recovery remains unclear. Herein, we investigated its therapeutic impact on a contusion model of moderate SCI. ALO (100 mg/kg/day) was intraperitoneally administered to adult Sprague-Dawley rats for a week following surgery/SCI. Basso-Beattie-Bresnahan locomotor score was used to assess neural function after post-SCI (day-1/4/7), showing that ALO modestly improved hind-limb locomotor recovery. HE-staining showed that ALO attenuated the increased tissue sparseness and liquefactive necrosis due to the contusion injury. ALO treatments reduced the injury-induced apoptosis (Bax/Bcl-2, cleaved-caspase3), oxidative (4HNE, MDA), and inflammatory (NF-κB, TNF-α) responses, and increased antioxidant enzymes SOD1 and GPx1 levels. The network pharmacology and immunoblot analyses revealed that the molecular targets of ALO and SCI include the PI3K/AKT pathway. Our findings, for the first time, clearly demonstrated that a natural compound, aloperine, has a neuroprotective effect on SCI by reducing apoptosis, inducing the antioxidant defense system, and modulating PI3K/AKT and NF-κB signaling. These results suggest that aloperine administration might improve the total antioxidant status and significantly promote functional recovery following traumatic SCI.
Collapse
Affiliation(s)
- Erhamit Okutan
- Neurosurgery Clinic, University of Health Sciences, Bağcılar Training and Research Hospital, Istanbul, Turkiye.
| | - İlker Güleç
- Neurosurgery Clinic, University of Health Sciences, Bağcılar Training and Research Hospital, Istanbul, Turkiye.
| | - Aslıhan Şengelen
- Department of Molecular Biology and Genetics, Faculty of Science, Istanbul University, Istanbul, Turkiye.
| | - Feyza Karagöz-Güzey
- Neurosurgery Clinic, University of Health Sciences, Bağcılar Training and Research Hospital, Istanbul, Turkiye.
| | - Burak Eren
- Neurosurgery Clinic, University of Health Sciences, Bağcılar Training and Research Hospital, Istanbul, Turkiye.
| | - Azmi Tufan
- Neurosurgery Clinic, University of Health Sciences, Bağcılar Training and Research Hospital, Istanbul, Turkiye.
| | - Tevhide Bilgen Özcan
- Department of Pathology, University of Health Sciences, Bağcılar Training and Research Hospital, Istanbul, Turkiye.
| | - Evren Önay-Uçar
- Department of Molecular Biology and Genetics, Faculty of Science, Istanbul University, Istanbul, Turkiye.
| |
Collapse
|
4
|
Wang YN, Liu S. The role of ALDHs in lipid peroxidation-related diseases. Int J Biol Macromol 2025; 288:138760. [PMID: 39674477 DOI: 10.1016/j.ijbiomac.2024.138760] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Revised: 11/26/2024] [Accepted: 12/11/2024] [Indexed: 12/16/2024]
Abstract
Lipid peroxidation presents the oxidative degradation of polyunsaturated fatty acids lincited by reactive species. Excessive accumulation of lipid peroxidation byproducts, including 4-hydroxy-2-nonenal (4-HNE) and malondialdehyde (MDA), causes protein dysfunction and various illnesses. Aldehyde dehydrogenases (ALDHs) catalyze the metabolism of both endogenous and exogenous aldehydes. These enzymes participate in detoxification and intermediary metabolism. Contemporary research has affirmed the involvement of both enzymatic and non-enzymatic pathways of ALDHs in modulating the evolution of diseases associated with lipid peroxidation. This review provides an overview of the biological functions and clinical implications concerning the enzymatic and non-enzymatic pathways of ALDHs in diseases related to lipid peroxidation, such as, non-alcoholic fatty liver disease (NAFLD), atherosclerosis, and type 2 diabetes (T2DM). Furthermore, the activators or inhibitors of ALDHs represent a promising therapeutic strategy for lipid peroxidation-related diseases.
Collapse
Affiliation(s)
- Ya-Nan Wang
- Department of Implantology & Periodontology, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Research Center of Dental Materials and Oral Tissue Regeneration & Shandong Provincial Clinical Research Center for Oral Diseases, Jinan, Shandong 250012, China; Suzhou Research Institute, Shandong University, Suzhou, Jiangsu 215123, China
| | - Shiyue Liu
- Department of Implantology & Periodontology, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Research Center of Dental Materials and Oral Tissue Regeneration & Shandong Provincial Clinical Research Center for Oral Diseases, Jinan, Shandong 250012, China.
| |
Collapse
|
5
|
Thorwald MA, Sta Maria NS, Chakhoyan A, O'Day PA, Jacobs RE, Zlokovic B, Finch CE. Iron chelation by oral deferoxamine treatment decreased brain iron and iron signaling proteins. J Alzheimers Dis 2025; 103:1180-1190. [PMID: 39894909 DOI: 10.1177/13872877241313031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2025]
Abstract
BACKGROUND Deferoxamine (DFO) and other iron chelators are clinically used for cancer and stroke. They may also be useful for Alzheimer's disease (AD) to diminish iron from microbleeds. DFO may also stimulate antioxidant membrane repair which is impaired during AD. DFO and other chelators do enter the brain despite some contrary reports. OBJECTIVE Low dose, oral DFO was given in lab chow to wildtype (WT) C57BL/6 mice to evaluate potential impact on iron levels, iron-signaling and storage proteins, and amyloid-β protein precursor (AβPP) and processing enzymes. Young WT mice do not have microbleeds or disrupted blood-brain barrier of AD mice. METHODS Iron was measured by MRI and chemically after two weeks of dietary DFO. Cerebral cortex was examined for changes in iron metabolism, antioxidant signaling, and AβPP processing by western blot. RESULTS DFO decreased brain iron 18% (p < 0.01) estimated by R2 MRI and decreased seven major proteins that mediate iron metabolism by at least 25%. The iron storage proteins ferritin light and heavy chain decreased by at least 30%. AβPP and secretase enzymes also decreased by 30%. CONCLUSIONS WT mice respond to DFO with decreased AβPP, amyloid processing enzymes, and antioxidant repair. Potential DFO treatment for early-stage AD by DFO should consider the benefits of lowered AβPP and secretase enzymes.
Collapse
Affiliation(s)
- Max A Thorwald
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA, USA
| | - Naomi S Sta Maria
- Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
- Department of Physiology and Neuroscience, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Ararat Chakhoyan
- Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
- Department of Physiology and Neuroscience, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Peggy A O'Day
- Life and Environmental Sciences Department, University of California, Merced, CA, USA
| | - Russell E Jacobs
- Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
- Department of Physiology and Neuroscience, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Berislav Zlokovic
- Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
- Department of Physiology and Neuroscience, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Caleb E Finch
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA, USA
- Dornsife College, University of Southern California, Los Angeles, CA, USA
| |
Collapse
|
6
|
Muhar MF, Farnung J, Cernakova M, Hofmann R, Henneberg LT, Pfleiderer MM, Denoth-Lippuner A, Kalčic F, Nievergelt AS, Peters Al-Bayati M, Sidiropoulos ND, Beier V, Mann M, Jessberger S, Jinek M, Schulman BA, Bode JW, Corn JE. C-terminal amides mark proteins for degradation via SCF-FBXO31. Nature 2025; 638:519-527. [PMID: 39880951 PMCID: PMC11821526 DOI: 10.1038/s41586-024-08475-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Accepted: 12/02/2024] [Indexed: 01/31/2025]
Abstract
During normal cellular homeostasis, unfolded and mislocalized proteins are recognized and removed, preventing the build-up of toxic byproducts1. When protein homeostasis is perturbed during ageing, neurodegeneration or cellular stress, proteins can accumulate several forms of chemical damage through reactive metabolites2,3. Such modifications have been proposed to trigger the selective removal of chemically marked proteins3-6; however, identifying modifications that are sufficient to induce protein degradation has remained challenging. Here, using a semi-synthetic chemical biology approach coupled to cellular assays, we found that C-terminal amide-bearing proteins (CTAPs) are rapidly cleared from human cells. A CRISPR screen identified FBXO31 as a reader of C-terminal amides. FBXO31 is a substrate receptor for the SKP1-CUL1-F-box protein (SCF) ubiquitin ligase SCF-FBXO31, which ubiquitylates CTAPs for subsequent proteasomal degradation. A conserved binding pocket enables FBXO31 to bind to almost any C-terminal peptide bearing an amide while retaining exquisite selectivity over non-modified clients. This mechanism facilitates binding and turnover of endogenous CTAPs that are formed after oxidative stress. A dominant human mutation found in neurodevelopmental disorders reverses CTAP recognition, such that non-amidated neosubstrates are now degraded and FBXO31 becomes markedly toxic. We propose that CTAPs may represent the vanguard of a largely unexplored class of modified amino acid degrons that could provide a general strategy for selective yet broad surveillance of chemically damaged proteins.
Collapse
Affiliation(s)
- Matthias F Muhar
- Institute of Molecular Health Sciences, Department of Biology, Swiss Federal Institute of Technology (ETH) Zurich, Zurich, Switzerland
| | - Jakob Farnung
- Laboratory for Organic Chemistry, Department of Chemistry and Applied Biosciences, Swiss Federal Institute of Technology (ETH) Zurich, Zurich, Switzerland
- Department of Molecular Machines and Signaling, Max Planck Institute of Biochemistry, Martinsried, Germany
| | - Martina Cernakova
- Institute of Molecular Health Sciences, Department of Biology, Swiss Federal Institute of Technology (ETH) Zurich, Zurich, Switzerland
| | - Raphael Hofmann
- Laboratory for Organic Chemistry, Department of Chemistry and Applied Biosciences, Swiss Federal Institute of Technology (ETH) Zurich, Zurich, Switzerland
| | - Lukas T Henneberg
- Department of Molecular Machines and Signaling, Max Planck Institute of Biochemistry, Martinsried, Germany
| | | | - Annina Denoth-Lippuner
- Laboratory of Neural Plasticity, Faculties of Medicine and Science, Brain Research Institute, University of Zurich, Zurich, Switzerland
| | - Filip Kalčic
- Laboratory for Organic Chemistry, Department of Chemistry and Applied Biosciences, Swiss Federal Institute of Technology (ETH) Zurich, Zurich, Switzerland
| | - Ajse S Nievergelt
- Department of Biochemistry, University of Zurich, Zurich, Switzerland
| | - Marwa Peters Al-Bayati
- Institute of Molecular Health Sciences, Department of Biology, Swiss Federal Institute of Technology (ETH) Zurich, Zurich, Switzerland
| | - Nikolaos D Sidiropoulos
- Institute of Molecular Health Sciences, Department of Biology, Swiss Federal Institute of Technology (ETH) Zurich, Zurich, Switzerland
| | - Viola Beier
- Department of Molecular Machines and Signaling, Max Planck Institute of Biochemistry, Martinsried, Germany
| | - Matthias Mann
- Department of Proteomics and Signal Transduction, Max Planck Institute of Biochemistry, Martinsried, Germany
| | - Sebastian Jessberger
- Laboratory of Neural Plasticity, Faculties of Medicine and Science, Brain Research Institute, University of Zurich, Zurich, Switzerland
| | - Martin Jinek
- Department of Biochemistry, University of Zurich, Zurich, Switzerland
| | - Brenda A Schulman
- Department of Molecular Machines and Signaling, Max Planck Institute of Biochemistry, Martinsried, Germany
| | - Jeffrey W Bode
- Laboratory for Organic Chemistry, Department of Chemistry and Applied Biosciences, Swiss Federal Institute of Technology (ETH) Zurich, Zurich, Switzerland.
| | - Jacob E Corn
- Institute of Molecular Health Sciences, Department of Biology, Swiss Federal Institute of Technology (ETH) Zurich, Zurich, Switzerland.
| |
Collapse
|
7
|
Otsuka Y, Yano M. FAM136A depletion induces mitochondrial stress and reduces mitochondrial membrane potential and ATP production. FEBS Open Bio 2025. [PMID: 39821719 DOI: 10.1002/2211-5463.13967] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Revised: 12/05/2024] [Accepted: 01/03/2025] [Indexed: 01/19/2025] Open
Abstract
FAM136A deficiency has been associated with Ménière's disease. However, the underlying mechanism of action of this protein remains unclear. We hypothesized that FAM136A functions in maintaining mitochondria, even in HepG2 cells. To better characterize FAM136A function, we analyzed the cellular response caused by its depletion. FAM136A depletion induced reactive oxygen species (ROS) and reduced both mitochondrial membrane potential and ATP production. However, cleaved caspase-9 levels did not increase significantly. We next investigated why the depletion of FAM136A reduced the mitochondrial membrane potential and ATP production but did not lead to apoptosis. Depletion of FAM136A induced the mitochondrial unfolded protein response (UPRmt) and the expression levels of gluconeogenic phosphoenolpyruvate carboxykinases (PCK1 and PCK2) and ketogenic 3-hydroxy-3-methylglutaryl-CoA synthases (HMGCS1 and HMGCS2) were upregulated. Furthermore, depletion of FAM136A reduced accumulation of holocytochrome c synthase (HCCS), a FAM136A interacting enzyme that combines heme to apocytochrome c to produce holocytochrome c. Notably, the amount of heme in cytochrome c did not change significantly with FAM136A depletion, although the amount of total cytochrome c protein increased significantly. This observation suggests that greater amounts of cytochrome c remain unbound to heme in FAM136A-depleted cells.
Collapse
Affiliation(s)
- Yushi Otsuka
- Department of Medical Technology, Faculty of Health Sciences, Kumamoto Health Science University, Kumamoto, Japan
| | - Masato Yano
- Department of Medical Technology, Faculty of Health Sciences, Kumamoto Health Science University, Kumamoto, Japan
| |
Collapse
|
8
|
Liu J, Zhong W, Wang R, Wang P, Tong G, Chai M, Sun Y, Zhu T, Huang C, Yang S, Zhou X, Mou D, Cai Y. Macrophage Ferroptotic Resistance Is Required for the Progression of Infantile Hemangioma. J Am Heart Assoc 2025; 14:e034261. [PMID: 39704244 DOI: 10.1161/jaha.124.034261] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/16/2024] [Accepted: 11/07/2024] [Indexed: 12/21/2024]
Abstract
BACKGROUND Ferroptosis is a programmed cell death caused by iron-dependent accumulation and cellular lipid peroxides, which is different from apoptosis and pyroptosis. This study investigated the possible effect of ferroptotic response in the pathogenesis of infantile hemangioma (IH). METHODS AND RESULTS The staining level of 4-hydroxynonenal (4-HNE), the marker of ferroptotic cells, was significantly increased in the involutive IH samples compared with the proliferative samples (9 proliferative versus 12 involutive lesions, P=0.0152). By contrast, the expression of glutathione peroxidase 4 (GPX4), a key enzyme regulating ferroptotic resistance, was significantly increased in the involutive IH samples. Meanwhile, the GPX4 was richly expressed in macrophages of IH. The data from in vitro study showed that the mRNA (P=0.0002) and protein (P=0.0385) expression levels of GPX4 were significantly upregulated in macrophages cultured with hemangioma-derived stem cells conditional medium (HemSC-CM). Mechanistically, HemSC-CM promoted the expression of GPX4 in macrophages (P=0.0482) by increasing nuclear factor erythroid 2-related factor 2 translocation to the nucleus (P=0.0026). Additionally, inhibition of GPX4 or inducing ferroptosis in macrophages could inhibit progression of lesion in IH nude mice mode. CONCLUSIONS Hemangioma-derived stem cells (HemSCs) could promote macrophage ferroptotic resistance through upregulating expression of GPX4, which is required for the progression of IH.
Collapse
Affiliation(s)
- Jingjing Liu
- State Key Laboratory of Oral and Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School and Hospital of Stomatology Wuhan University Wuhan China
- Department of Oral and Maxillofacial Surgery, School and Hospital of Stomatology Wuhan University Wuhan China
| | - Wenqun Zhong
- State Key Laboratory of Oral and Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School and Hospital of Stomatology Wuhan University Wuhan China
- Department of Oral and Maxillofacial Surgery, School and Hospital of Stomatology Wuhan University Wuhan China
| | - Rong Wang
- State Key Laboratory of Oral and Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School and Hospital of Stomatology Wuhan University Wuhan China
| | - Peipei Wang
- State Key Laboratory of Oral and Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School and Hospital of Stomatology Wuhan University Wuhan China
- Department of Oral and Maxillofacial Surgery, School and Hospital of Stomatology Wuhan University Wuhan China
| | - Guoyong Tong
- Department of Stomatology The Central Hospital of Enshi Tujia and Miao Autonomous Perfecture Enshi China
| | - Maosheng Chai
- State Key Laboratory of Oral and Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School and Hospital of Stomatology Wuhan University Wuhan China
- Department of Oral and Maxillofacial Surgery, School and Hospital of Stomatology Wuhan University Wuhan China
| | - Yu Sun
- Department of Plastic Surgery Wuhan Children's Hospital Wuhan China
| | - Tianshuang Zhu
- State Key Laboratory of Oral and Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School and Hospital of Stomatology Wuhan University Wuhan China
- Department of Oral and Maxillofacial Surgery, School and Hospital of Stomatology Wuhan University Wuhan China
| | - Congfa Huang
- State Key Laboratory of Oral and Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School and Hospital of Stomatology Wuhan University Wuhan China
- Department of Oral and Maxillofacial Surgery, School and Hospital of Stomatology Wuhan University Wuhan China
| | - Shaodong Yang
- Department of Pathology, School and Hospital of Stomatology Wuhan University Wuhan China
| | | | | | - Yu Cai
- State Key Laboratory of Oral and Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School and Hospital of Stomatology Wuhan University Wuhan China
- Department of Oral and Maxillofacial Surgery, School and Hospital of Stomatology Wuhan University Wuhan China
| |
Collapse
|
9
|
Wang Y, Zhu S, He W, Marchuk H, Richard E, Desviat LR, Young SP, Koeberl D, Kasumov T, Chen X, Zhang GF. The attenuated hepatic clearance of propionate increases cardiac oxidative stress in propionic acidemia. Basic Res Cardiol 2024; 119:1045-1062. [PMID: 38992300 PMCID: PMC11702364 DOI: 10.1007/s00395-024-01066-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/17/2023] [Revised: 06/29/2024] [Accepted: 06/30/2024] [Indexed: 07/13/2024]
Abstract
Propionic acidemia (PA), arising from PCCA or PCCB variants, manifests as life-threatening cardiomyopathy and arrhythmias, with unclear pathophysiology. In this work, propionyl-CoA metabolism in rodent hearts and human pluripotent stem cell-derived cardiomyocytes was investigated with stable isotope tracing analysis. Surprisingly, gut microbiome-derived propionate rather than the propiogenic amino acids (valine, isoleucine, threonine, and methionine) or odd-chain fatty acids was found to be the primary cardiac propionyl-CoA source. In a Pcca-/-(A138T) mouse model and PA patients, accumulated propionyl-CoA and diminished acyl-CoA synthetase short-chain family member 3 impede hepatic propionate disposal, elevating circulating propionate. Prolonged propionate exposure induced significant oxidative stress in PCCA knockdown HL-1 cells and the hearts of Pcca-/-(A138T) mice. Additionally, Pcca-/-(A138T) mice exhibited mild diastolic dysfunction after the propionate challenge. These findings suggest that elevated circulating propionate may cause oxidative damage and functional impairment in the hearts of patients with PA.
Collapse
Affiliation(s)
- You Wang
- School of Basic Medicine, Jining Medical University, Shandong, 272067, China
- Duke Molecular Physiology Institute and Sarah W. Stedman Nutrition and Metabolism Center, Duke University Medical Center, Carmichael Building 48-203, 300 North Duke Street, Durham, NC, 27701, USA
| | - Suhong Zhu
- School of Basic Medicine, Jining Medical University, Shandong, 272067, China
- Duke Molecular Physiology Institute and Sarah W. Stedman Nutrition and Metabolism Center, Duke University Medical Center, Carmichael Building 48-203, 300 North Duke Street, Durham, NC, 27701, USA
| | - Wentao He
- Duke Molecular Physiology Institute and Sarah W. Stedman Nutrition and Metabolism Center, Duke University Medical Center, Carmichael Building 48-203, 300 North Duke Street, Durham, NC, 27701, USA
| | - Hannah Marchuk
- Duke Molecular Physiology Institute and Sarah W. Stedman Nutrition and Metabolism Center, Duke University Medical Center, Carmichael Building 48-203, 300 North Duke Street, Durham, NC, 27701, USA
| | - Eva Richard
- Centro de Biología Molecular Severo Ochoa UAM-CSIC, CIBERER, IdiPaz, IUBM, Universidad Autónoma de Madrid, Madrid, Spain
| | - Lourdes R Desviat
- Centro de Biología Molecular Severo Ochoa UAM-CSIC, CIBERER, IdiPaz, IUBM, Universidad Autónoma de Madrid, Madrid, Spain
| | - Sarah P Young
- Biochemical Genetics Laboratory, Duke University Health System, Durham, NC, USA
- Division of Medical Genetics, Department of Pediatrics, Duke University School of Medicine, Duke University Medical Center, Durham, NC, 27710, USA
| | - Dwight Koeberl
- Division of Medical Genetics, Department of Pediatrics, Duke University School of Medicine, Duke University Medical Center, Durham, NC, 27710, USA
| | - Takhar Kasumov
- Department of Pharmaceutical Sciences, College of Pharmacy, Northeast Ohio Medical University, Rootstown, OH, USA
| | - Xiaoxin Chen
- Surgical Research Lab, Department of Surgery, Cooper University Hospital and Cooper Medical School of Rowan University, Camden, NJ, 08103, USA
- Coriell Institute for Medical Research, Camden, NJ, 08103, USA
- MD Anderson Cancer Center at Cooper, Camden, NJ, 08103, USA
| | - Guo-Fang Zhang
- Duke Molecular Physiology Institute and Sarah W. Stedman Nutrition and Metabolism Center, Duke University Medical Center, Carmichael Building 48-203, 300 North Duke Street, Durham, NC, 27701, USA.
- Department of Medicine, Division of Endocrinology, Metabolism and Nutrition, Duke University Medical Center, Durham, NC, 27701, USA.
| |
Collapse
|
10
|
Kargı-Gemici E, Şengelen A, Aksüt Y, Akyol O, Şengiz-Erhan S, Bay M, Önay-Uçar E, Selcan A, Demirgan S. Cerium oxide nanoparticles (nanoceria) pretreatment attenuates cell death in the hippocampus and cognitive dysfunction due to repeated isoflurane anesthesia in newborn rats. Neurotoxicology 2024; 105:82-93. [PMID: 39216603 DOI: 10.1016/j.neuro.2024.08.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2024] [Revised: 08/15/2024] [Accepted: 08/26/2024] [Indexed: 09/04/2024]
Abstract
General anesthetics exposure, particularly prolonged or repeated exposure, is a crucial cause of neurological injuries. Notably, isoflurane (ISO), used in pediatric anesthesia practice, is toxic to the developing brain. The relatively weak antioxidant system at early ages needs antioxidant support to protect the brain against anesthesia. Cerium oxide nanoparticles (CeO2-NPs, nanoceria) are nano-antioxidants and stand out due to their unique surface chemistry, high stability, and biocompatibility. Although CeO2-NPs have been shown to exhibit neuroprotective and cognitive function-facilitating effects, there are no reports on their protective effects against anesthesia-induced neurotoxicity and cognitive impairments. Herein, Wistar albino rat pups were exposed to ISO (1.5 %, 3-h) at postnatal day (P)7+P9+P11, and the protective properties of CeO2-NP pretreatment (0.5 mg/kg, intraperitoneal route) were investigated for the first time. The control group at P7+9+11 received 50 % O2 (3-h) instead of ISO. Exposure to nanoceria one-hour before ISO protected hippocampal neurons of the developing rat brain against apoptosis [determined by hematoxylin-eosin (HE) staining, immunohistochemistry (IHC) analysis with caspase-3, and immunoblotting with Bax/Bcl2, cleaved caspase-3 and PARP1] oxidative stress, and inflammation [determined by immunoblotting with 4-hydroxynonenal (4HNE), nuclear factor kappa-B (NF-κB), and tumor necrosis factor-alpha (TNF-α)]. CeO2-NP pretreatment also reduced ISO-induced learning (at P28-32) and memory (at P33) deficits evaluated by Morris Water Maze. However, memory deficits and thigmotactic behaviors were detected in the agent-control group; elimination of these harmful effects will be possible with dose studies, thus providing evidence supporting safer use. Overall, our findings support pretreatment with nanoceria application as a simple strategy that might be used for pediatric anesthesia practice to protect infants and children from ISO-induced cell death and learning and memory deficits.
Collapse
Affiliation(s)
- Ezgi Kargı-Gemici
- Clinic of Anesthesiology and Reanimation, University of Health Sciences, Bağcılar Training and Research Hospital, Istanbul, Turkiye.
| | - Aslıhan Şengelen
- Department of Molecular Biology and Genetics, Faculty of Science, Istanbul University, Istanbul, Turkiye.
| | - Yunus Aksüt
- Department of Molecular Biology and Genetics, Faculty of Science, Istanbul University, Istanbul, Turkiye; Department of Molecular Biology and Genetics, Basic Medical Sciences, School of Medicine, Koç University, Istanbul, Turkiye.
| | - Onat Akyol
- Clinic of Anesthesiology and Reanimation, Istanbul Florence Nightingale Hospital, Istanbul, Turkiye.
| | - Selma Şengiz-Erhan
- Clinic of Pathology, University of Health Sciences, Prof. Dr. Cemil Taşçıoğlu City Hospital, Istanbul, Turkiye.
| | - Mehmet Bay
- Clinic of Anesthesiology and Reanimation, University of Health Sciences, Bağcılar Training and Research Hospital, Istanbul, Turkiye.
| | - Evren Önay-Uçar
- Department of Molecular Biology and Genetics, Faculty of Science, Istanbul University, Istanbul, Turkiye.
| | - Ayşin Selcan
- Clinic of Anesthesiology and Reanimation, University of Health Sciences, Bağcılar Training and Research Hospital, Istanbul, Turkiye.
| | - Serdar Demirgan
- Clinic of Anesthesiology and Reanimation, University of Health Sciences, Bağcılar Training and Research Hospital, Istanbul, Turkiye; Department of Molecular Biology and Genetics, Institute of Graduate Studies in Sciences, Istanbul University, Istanbul, Turkiye.
| |
Collapse
|
11
|
De Rubeis M, Mascitti IA, Cocciolone D, Placidi M, Vergara T, Di Emidio G, Macchiarelli G, Tatone C, Nottola SA, Palmerini MG. Morphological and Redox/Glycative Alterations in the PCOS Oviducts: Modulating Effects of Carnitines in PCOS Mice. BIOLOGY 2024; 13:964. [PMID: 39765631 PMCID: PMC11673334 DOI: 10.3390/biology13120964] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Revised: 11/21/2024] [Accepted: 11/22/2024] [Indexed: 01/11/2025]
Abstract
Polycystic ovarian syndrome (PCOS) is a heterogeneous condition characterized by hyperandrogenism (HA), polycystic ovaries, and dysfunctional ovulation, and it is associated with metabolic problems such as insulin resistance (IR) and obesity. After having investigated the morphological and antioxidant/antiglycative alterations on mouse ovaries and uteri, we here focus on PCOS oviducts, a tract of the reproductive system essential for the nourishment and transport of gametes and embryos. The modulating effects of L-carnitine (LC) and acetyl-L-carnitine (ALC) were also assessed. CD1 mice were administered or not with dehydroepiandrosterone (DHEA, 6 mg/100 g body weight) for 20 days, alone or with 0.40 mg of L-carnitine (LC) and 0.20 mg of acetyl-L-carnitine (ALC). Oviducts were then subjected to histology and immunohistochemistry to evaluate their morphology and collagen deposition, and steroidogenesis. Oxidative, mitochondrial, and methylglyoxal (MG)-dependent damage was also investigated. Transmission electron microscopy was used to detect ultrastructural alterations. The PCOS oviducts were affected by hyperfibrosis, hyperplasia, hypertrophy, and altered steroidogenesis, with oxidative alterations associated with MethylGlyoxal-Advanced Glycation End product (MG-AGE) accumulation. A reduced ciliary coverage and numerous dilated intercellular spaces were found in the epithelium. LC-ALC administration mitigated PCOS oviductal alterations. These results provide evidence for the detrimental action of oxidative and glycative stress in PCOS oviducts, confirming a protective role of carnitines on the PCOS phenotype.
Collapse
Affiliation(s)
- Mariacarla De Rubeis
- Department of Anatomy, Histology, Forensic Medicine and Orthopaedics, Sapienza University, 00161 Rome, Italy; (M.D.R.); (S.A.N.)
| | - Ilaria Antenisca Mascitti
- Department of Life, Health and Environmental Sciences, University of L’Aquila, 67100 L’Aquila, Italy; (I.A.M.); (D.C.); (M.P.); (T.V.); (G.D.E.); (G.M.); (C.T.)
| | - Domenica Cocciolone
- Department of Life, Health and Environmental Sciences, University of L’Aquila, 67100 L’Aquila, Italy; (I.A.M.); (D.C.); (M.P.); (T.V.); (G.D.E.); (G.M.); (C.T.)
| | - Martina Placidi
- Department of Life, Health and Environmental Sciences, University of L’Aquila, 67100 L’Aquila, Italy; (I.A.M.); (D.C.); (M.P.); (T.V.); (G.D.E.); (G.M.); (C.T.)
| | - Teresa Vergara
- Department of Life, Health and Environmental Sciences, University of L’Aquila, 67100 L’Aquila, Italy; (I.A.M.); (D.C.); (M.P.); (T.V.); (G.D.E.); (G.M.); (C.T.)
| | - Giovanna Di Emidio
- Department of Life, Health and Environmental Sciences, University of L’Aquila, 67100 L’Aquila, Italy; (I.A.M.); (D.C.); (M.P.); (T.V.); (G.D.E.); (G.M.); (C.T.)
| | - Guido Macchiarelli
- Department of Life, Health and Environmental Sciences, University of L’Aquila, 67100 L’Aquila, Italy; (I.A.M.); (D.C.); (M.P.); (T.V.); (G.D.E.); (G.M.); (C.T.)
| | - Carla Tatone
- Department of Life, Health and Environmental Sciences, University of L’Aquila, 67100 L’Aquila, Italy; (I.A.M.); (D.C.); (M.P.); (T.V.); (G.D.E.); (G.M.); (C.T.)
| | - Stefania Annarita Nottola
- Department of Anatomy, Histology, Forensic Medicine and Orthopaedics, Sapienza University, 00161 Rome, Italy; (M.D.R.); (S.A.N.)
| | - Maria Grazia Palmerini
- Department of Life, Health and Environmental Sciences, University of L’Aquila, 67100 L’Aquila, Italy; (I.A.M.); (D.C.); (M.P.); (T.V.); (G.D.E.); (G.M.); (C.T.)
| |
Collapse
|
12
|
Alhegaili AS, Bafail DA, Bawahab AA, Alsubaie N, Abd-Elhakim YM, Mohamed AAR, Khamis T, Khalifa NE, Elhamouly M, Dahran N, El Shetry ES. The interplay of oxidative stress, apoptotic signaling, and impaired mitochondrial function in the pyrethroid-induced cardiac injury: Alleviative role of curcumin-loaded chitosan nanoparticle. Food Chem Toxicol 2024; 194:115095. [PMID: 39515510 DOI: 10.1016/j.fct.2024.115095] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Revised: 11/04/2024] [Accepted: 11/05/2024] [Indexed: 11/16/2024]
Abstract
This study assessed the consequence of exposure to a pyrethroid insecticide, fenpropathrin (FPN), on the heart and the probable underlying mechanisms in rats. Moreover, the probable protective effect of curcumin-loaded chitosan nanoparticles (CMN-CNP) was evaluated. Forty male Sprague Dawley rats were distributed into four groups orally given corn oil, CMN-CNP (50 mg/kg b.wt), FPN (15 mg/kg b.wt), or CMN-CNP + FPN for 60 days. The results revealed that FPN exposure increased serum cardiac damage indicators. In addition, a substantial increase in the reactive oxygen species and malondialdehyde content but reduced enzymatic and non-enzymatic antioxidants and altered architecture was recorded in the cardiac tissue of FPN-exposed rats. Additionally, a significant down-regulation of expression of the mitochondrial complexes I-V, mitochondrial dynamics, and antioxidants-related genes but up-regulation of apoptosis-related genes was detected in the FPN-exposed group. Immunofluorescence analyses revealed higher amounts of the harmful protein 4-hydroxynonenal in the heart tissue of FPN-exposed rats. Nevertheless, the earlier disturbances were significantly rescued in the FPN + CMN-CNP treated group. Conclusively, our findings reported the cardiotoxic activity of FPN and the involvement of several mitochondrial imbalances as a probable underlying mechanism. Also, the study findings proved the efficacy of CMN-CNP in combating FPN cardiotoxic effects.
Collapse
Affiliation(s)
- Alaa S Alhegaili
- Department of Medical Laboratory, College of Applied Medical Sciences, Prince Sattam Bin Abdulaziz University, Al-Kharj 11942, Saudi Arabia
| | - Duaa Abdullah Bafail
- Department of Clinical Pharmacology, Faculty of Medicine, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Ahmed Abdulwahab Bawahab
- Department of Basic Medical Sciences, College of Medicine, University of Jeddah, Jeddah, Saudi Arabia
| | - Nawal Alsubaie
- Department of Pharmacy Practice, College of Pharmacy, Princess Nourah bint Abdulrahman University, P.O. Box 84428, Riyadh 11671, Saudi Arabia.
| | - Yasmina M Abd-Elhakim
- Department of Forensic Medicine and Toxicology, Faculty of Veterinary Medicine, Zagazig University, Zagazig 44519, Egypt
| | - Amany Abdel-Rahman Mohamed
- Department of Forensic Medicine and Toxicology, Faculty of Veterinary Medicine, Zagazig University, Zagazig 44519, Egypt
| | - Tarek Khamis
- Department of Pharmacology, Faculty of Veterinary Medicine, Zagazig University, Zagazig 44519, Egypt; Laboratory of Biotechnology, Faculty of Veterinary Medicine, Zagazig University, Zagazig 44519, Egypt
| | - Norhan E Khalifa
- Department of Physiology, Faculty of Veterinary Medicine, Matrouh University, Matrouh 51511, Egypt
| | - Moustafa Elhamouly
- Department of Histology and Cytology Faculty of Veterinary Medicine, University of Sadat City, Sadat City, Egypt
| | - Naief Dahran
- Department of Basic Medical Sciences, College of Medicine, University of Jeddah, Jeddah, Saudi Arabia
| | - Eman S El Shetry
- Department of Anatomy, College of Medicine, University of Hail, Hail, Saudi Arabia; Department of Human Anatomy and Embryology, Faculty of Medicine, Zagazig University, Zagazig 44519, Egypt
| |
Collapse
|
13
|
Atalay Ekiner S, Gęgotek A, Skrzydlewska E. Inflammasome activity regulation by PUFA metabolites. Front Immunol 2024; 15:1452749. [PMID: 39290706 PMCID: PMC11405227 DOI: 10.3389/fimmu.2024.1452749] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Accepted: 08/19/2024] [Indexed: 09/19/2024] Open
Abstract
Oxidative stress and the accompanying chronic inflammation constitute an important metabolic problem that may lead to pathology, especially when the body is exposed to physicochemical and biological factors, including UV radiation, pathogens, drugs, as well as endogenous metabolic disorders. The cellular response is associated, among others, with changes in lipid metabolism, mainly due to the oxidation and the action of lipolytic enzymes. Products of oxidative fragmentation/cyclization of polyunsaturated fatty acids (PUFAs) [4-HNE, MDA, 8-isoprostanes, neuroprostanes] and eicosanoids generated as a result of the enzymatic metabolism of PUFAs significantly modify cellular metabolism, including inflammation and the functioning of the immune system by interfering with intracellular molecular signaling. The key regulators of inflammation, the effectiveness of which can be regulated by interacting with the products of lipid metabolism under oxidative stress, are inflammasome complexes. An example is both negative or positive regulation of NLRP3 inflammasome activity by 4-HNE depending on the severity of oxidative stress. 4-HNE modifies NLRP3 activity by both direct interaction with NLRP3 and alteration of NF-κB signaling. Furthermore, prostaglandin E2 is known to be positively correlated with both NLRP3 and NLRC4 activity, while its potential interference with AIM2 or NLRP1 activity is unproven. Therefore, the influence of PUFA metabolites on the activity of well-characterized inflammasome complexes is reviewed.
Collapse
Affiliation(s)
| | - Agnieszka Gęgotek
- Department of Analytical Chemistry, Medical University of Bialystok, Bialystok, Poland
| | - Elżbieta Skrzydlewska
- Department of Analytical Chemistry, Medical University of Bialystok, Bialystok, Poland
| |
Collapse
|
14
|
George J, Lu Y, Tsuchishima M, Tsutsumi M. Cellular and molecular mechanisms of hepatic ischemia-reperfusion injury: The role of oxidative stress and therapeutic approaches. Redox Biol 2024; 75:103258. [PMID: 38970988 PMCID: PMC11279328 DOI: 10.1016/j.redox.2024.103258] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Revised: 06/20/2024] [Accepted: 06/25/2024] [Indexed: 07/08/2024] Open
Abstract
Ischemia-reperfusion (IR) or reoxygenation injury is the paradoxical exacerbation of cellular impairment following restoration of blood flow after a period of ischemia during surgical procedures or other conditions. Acute interruption of blood supply to the liver and subsequent reperfusion can result in hepatocyte injury, apoptosis, and necrosis. Since the liver requires a continuous supply of oxygen for many biochemical reactions, any obstruction of blood flow can rapidly lead to hepatic hypoxia, which could quickly progress to absolute anoxia. Reoxygenation results in the increased generation of reactive oxygen species and oxidative stress, which lead to the enhanced production of proinflammatory cytokines, chemokines, and other signaling molecules. Consequent acute inflammatory cascades lead to significant impairment of hepatocytes and nonparenchymal cells. Furthermore, the expression of several vascular growth factors results in the heterogeneous closure of numerous hepatic sinusoids, which leads to reduced oxygen supply in certain areas of the liver even after reperfusion. Therefore, it is vital to identify appropriate therapeutic modalities to mitigate hepatic IR injury and subsequent tissue damage. This review covers all the major aspects of cellular and molecular mechanisms underlying the pathogenesis of hepatic ischemia-reperfusion injury, with special emphasis on oxidative stress, associated inflammation and complications, and prospective therapeutic approaches.
Collapse
Affiliation(s)
- Joseph George
- Department of Cancer Biology, Mayo Clinic, Jacksonville, FL, 32224, USA; Department of Hepatology, Kanazawa Medical University, Uchinada, Ishikawa, 920-0293, Japan; Center for Regenerative Medicine, Kanazawa Medical University Hospital, Uchinada, Ishikawa, 920-0293, Japan.
| | - Yongke Lu
- Department of Biomedical Sciences, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV, 25755, USA
| | - Mutsumi Tsuchishima
- Department of Hepatology, Kanazawa Medical University, Uchinada, Ishikawa, 920-0293, Japan
| | - Mikihiro Tsutsumi
- Department of Hepatology, Kanazawa Medical University, Uchinada, Ishikawa, 920-0293, Japan; Center for Regenerative Medicine, Kanazawa Medical University Hospital, Uchinada, Ishikawa, 920-0293, Japan
| |
Collapse
|
15
|
Chen KW, Chen YR, Yang LY, Cheng YW, Chou SC, Chen YH, Chen YT, Hsieh ST, Kuo MF, Wang KC. Microcirculatory Impairment and Cerebral Injury in Hydrocephalus and the Effects of Cerebrospinal Fluid Diversion. Neurosurgery 2024; 95:469-479. [PMID: 38511941 DOI: 10.1227/neu.0000000000002908] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Accepted: 01/22/2024] [Indexed: 03/22/2024] Open
Abstract
BACKGROUND AND OBJECTIVES Hydrocephalus is characterized by progressive enlargement of cerebral ventricles, resulting in impaired microvasculature and cerebral hypoperfusion. This study aimed to demonstrate the microvascular changes in hydrocephalic rats and the effects of cerebrospinal fluid (CSF) release on cerebral blood flow (CBF). METHODS On postnatal day 21 (P21), male Wistar rats were intracisternally injected with either a kaolin suspension or saline. On P47, Evan's ratio (ER) was measured using MRI. On P49, the arteriolar diameter and vascular density of the pia were quantified using a capillary video microscope. The CBF was measured using laser Doppler flowmetry. The expressions of NeuN and glial fibrillary acidic protein determined by immunochemical staining were correlated with the ER. The CBF and rotarod test performance were recorded before and after CSF release. The expressions of 4-hydroxynonenal (4-HNE) and c-caspase-3 were studied on P56. RESULTS Ventriculomegaly was induced to varying degrees, resulting in the stretching and abnormal narrowing of pial arterioles, which regressed with increasing ER. Quantitative analysis revealed significant decreases in the arteriolar diameter and vascular density in the hydrocephalic group compared with those in the control group. In addition, the CBF in the hydrocephalic group decreased to 30%-50% of that in the control group. In hydrocephalus, the neurons appear distorted, and the expression of 4-HNE and reactive astrogliosis increase in the cortex. After CSF was released, improvements in the CBF and rotarod test performance were inversely associated with the ER. In addition, the levels of 4-HNE and c-caspase-3 were further elevated. CONCLUSION Rapid ventricular dilatation is associated with severe microvascular distortion, vascular regression, cortical hypoperfusion, and cellular changes that impair the recovery of CBF and motor function after CSF release. Moreover, CSF release may induce reperfusion injury. This pathophysiology should be taken into account when treating hydrocephalus.
Collapse
Affiliation(s)
- Kuo-Wei Chen
- Division of Neurosurgery, Department of Surgery, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei , Taiwan
- Graduate Institute of Clinical Medicine, National Taiwan University College of Medicine, Taipei , Taiwan
- Master Degree of Public Health, College of Public Health, National Taiwan University, Taipei , Taiwan
| | - Yong-Ren Chen
- Non-invasive Cancer Therapy Research Institute, Taipei , Taiwan
| | - Ling-Yu Yang
- Division of Neurosurgery, Department of Surgery, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei , Taiwan
| | - Ya-Wen Cheng
- Division of Neurosurgery, Department of Surgery, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei , Taiwan
| | - Sheng-Che Chou
- Division of Neurosurgery, Department of Surgery, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei , Taiwan
- Graduate Institute of Clinical Medicine, National Taiwan University College of Medicine, Taipei , Taiwan
- Department of Traumatology, National Taiwan University Hospital, National Taiwan University, College of Medicine, Taipei , Taiwan
| | - Yi-Hsing Chen
- Division of Neurosurgery, Department of Surgery, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei , Taiwan
- Graduate Institute of Clinical Medicine, National Taiwan University College of Medicine, Taipei , Taiwan
| | - Yi-Tzu Chen
- Division of Neurosurgery, Department of Surgery, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei , Taiwan
| | - Sung-Tsang Hsieh
- Department of Neurology, National Taiwan University Hospital, Taipei , Taiwan
- Department of Anatomy and Cell Biology, National Taiwan University College of Medicine, Taipei , Taiwan
| | - Meng-Fai Kuo
- Division of Neurosurgery, Department of Surgery, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei , Taiwan
| | - Kuo-Chuan Wang
- Division of Neurosurgery, Department of Surgery, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei , Taiwan
| |
Collapse
|
16
|
Zhong T, Li Y, Jin M, Liu J, Wu Z, Zhu F, Zhao L, Fan Y, Xu L, Ji J. Downregulation of 4-HNE and FOXO4 collaboratively promotes NSCLC cell migration and tumor growth. Cell Death Dis 2024; 15:546. [PMID: 39085238 PMCID: PMC11291900 DOI: 10.1038/s41419-024-06948-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2024] [Revised: 07/15/2024] [Accepted: 07/24/2024] [Indexed: 08/02/2024]
Abstract
Non-small cell lung cancer (NSCLC) is among the most prevalent cancers and a leading cause of cancer-related mortality globally. Extracellular vesicles (EVs) derived from NSCLC play a pivotal role in lung cancer progression. Our findings reveal a direct correlation between the abundance of EVs and the transfection efficiencies. Co-culturing two different lung cancer cell lines could enhance EVs formation, cell proliferation, migration and tumorigenicity. mRNA chip and metabolic analyses revealed significant alterations in the FOXO signaling pathway and unsaturated fatty acid metabolism within tumor tissues derived from co-cultured cells. Shotgun lipidomics studies and bioinformatics analyses guided our attention towards 4-Hydroxynonenal (4-HNE) and FOXO4. Elevating 4-HNE or FOXO4 levels could reduce the formation of EVs and impede cell growth and migration. While silencing FOXO4 expression lead to an increase in cell cloning rate and enhanced migration. These findings suggest that regulating the production of 4-HNE and FOXO4 might provide an effective therapeutic approach for the treatment of NSCLC.
Collapse
Affiliation(s)
- Tianfei Zhong
- College of Basic Medical, Zhejiang Chinese Medical University, Hangzhou, China
- Logistic Affairs Department, Zhejiang Chinese Medical University, Hangzhou, China
| | - Ying Li
- The First School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, China
| | - Meng Jin
- The First School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, China
| | - Jingqun Liu
- The First School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, China
| | - Zhenyu Wu
- College of Basic Medical, Zhejiang Chinese Medical University, Hangzhou, China
- Key Laborat Laboratory of Chinese Medicine Rtheumatology of Zhejiang Province, Hangzhou, China
| | - Feiye Zhu
- Academy of Chinese Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Lisha Zhao
- Department of Medicine, Zhejiang Academy of Traditional Chinese Medicine, Hangzhou, China
| | - Yongsheng Fan
- Key Laborat Laboratory of Chinese Medicine Rtheumatology of Zhejiang Province, Hangzhou, China
- Department of Rheumatology, The Second Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China
| | - Li Xu
- College of Basic Medical, Zhejiang Chinese Medical University, Hangzhou, China.
- Key Laborat Laboratory of Chinese Medicine Rtheumatology of Zhejiang Province, Hangzhou, China.
| | - Jinjun Ji
- College of Basic Medical, Zhejiang Chinese Medical University, Hangzhou, China.
- Key Laborat Laboratory of Chinese Medicine Rtheumatology of Zhejiang Province, Hangzhou, China.
| |
Collapse
|
17
|
Lin D, Gold A, Kaye S, Atkinson JR, Tol M, Sas A, Segal B, Tontonoz P, Zhu J, Gao J. Arachidonic Acid Mobilization and Peroxidation Promote Microglial Dysfunction in Aβ Pathology. J Neurosci 2024; 44:e0202242024. [PMID: 38866484 PMCID: PMC11293449 DOI: 10.1523/jneurosci.0202-24.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Revised: 05/07/2024] [Accepted: 05/29/2024] [Indexed: 06/14/2024] Open
Abstract
Aberrant increase of arachidonic acid (ARA) has long been implicated in the pathology of Alzheimer's disease (AD), while the underlying causal mechanism remains unclear. In this study, we revealed a link between ARA mobilization and microglial dysfunction in Aβ pathology. Lipidomic analysis of primary microglia from AppNL-GF mice showed a marked increase in free ARA and lysophospholipids (LPLs) along with a decrease in ARA-containing phospholipids, suggesting increased ARA release from phospholipids (PLs). To manipulate ARA-containing PLs in microglia, we genetically deleted lysophosphatidylcholine acyltransferase 3 (Lpcat3), the main enzyme catalyzing the incorporation of ARA into PLs. Loss of microglial Lpcat3 reduced the levels of ARA-containing PLs, free ARA and LPLs, leading to a compensatory increase in monounsaturated fatty acid (MUFA)-containing PLs in both male and female App NL-GF mice. Notably, the reduction of ARA in microglia significantly ameliorated oxidative stress and inflammatory responses while enhancing the phagocytosis of Aβ plaques and promoting the compaction of Aβ deposits. Mechanistically, scRNA seq suggested that LPCAT3 deficiency facilitates phagocytosis by facilitating de novo lipid synthesis while protecting microglia from oxidative damage. Collectively, our study reveals a novel mechanistic link between ARA mobilization and microglial dysfunction in AD. Lowering brain ARA levels through pharmacological or dietary interventions may be a potential therapeutic strategy to slow down AD progression.
Collapse
Affiliation(s)
- Da Lin
- Department of Neuroscience, The Ohio State University Wexner Medical Center, Columbus, Ohio 43210
| | - Andrew Gold
- Department of Human Sciences, The Ohio State University, Columbus, Ohio 43210
| | - Sarah Kaye
- Department of Neuroscience, The Ohio State University Wexner Medical Center, Columbus, Ohio 43210
| | - Jeffrey R Atkinson
- Department of Neurology, The Ohio State University Wexner Medical Center, Columbus, Ohio 43210
| | - Marcus Tol
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California, Los Angeles, California 90095
| | - Andrew Sas
- Department of Neurology, The Ohio State University Wexner Medical Center, Columbus, Ohio 43210
| | - Benjamin Segal
- Department of Neurology, The Ohio State University Wexner Medical Center, Columbus, Ohio 43210
| | - Peter Tontonoz
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California, Los Angeles, California 90095
| | - Jiangjiang Zhu
- Department of Human Sciences, The Ohio State University, Columbus, Ohio 43210
| | - Jie Gao
- Department of Neuroscience, The Ohio State University Wexner Medical Center, Columbus, Ohio 43210
| |
Collapse
|
18
|
Kültz D, Gardell AM, DeTomaso A, Stoney G, Rinkevich B, Qarri A, Hamar J. Proteome-wide 4-hydroxy-2-nonenal signature of oxidative stress in the marine invasive tunicate Botryllus schlosseri. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.19.604351. [PMID: 39211222 PMCID: PMC11360967 DOI: 10.1101/2024.07.19.604351] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/04/2024]
Abstract
The colonial ascidian Boytryllus schlosseri is an invasive marine chordate that thrives under conditions of anthropogenic climate change. We show that the B. schlosseri expressed proteome contains unusually high levels of proteins that are adducted with 4-hydroxy-2-nonenal (HNE). HNE represents a prominent posttranslational modification resulting from oxidative stress. Although numerous studies have assessed oxidative stress in marine organisms HNE protein modification has not previously been determined in any marine species. LC/MS proteomics was used to identify 1052 HNE adducted proteins in B. schlosseri field and laboratory populations. Adducted amino acid residues were ascertained for 1849 modified sites, of which 1195 had a maximum amino acid localization score. Most HNE modifications were at less reactive lysines (rather than more reactive cysteines). HNE prevelance on most sites was high. These observations suggest that B. schlosseri experiences and tolerates high intracellular reactive oxygen species levels, resulting in substantial lipid peroxidation. HNE adducted B. schlosseri proteins show enrichment in mitochondrial, proteostasis, and cytoskeletal functions. Based on these results we propose that redox signaling contributes to regulating energy metabolism, the blastogenic cycle, oxidative burst defenses, and cytoskeleton dynamics during B. schlosseri development and physiology. A DIA assay library was constructed to quantify HNE adduction at 72 sites across 60 proteins that represent a holistic network of functionally discernable oxidative stress bioindicators. We conclude that the vast amount of HNE protein adduction in this circumpolar tunicate is indicative of high oxidative stress tolerance contributing to its range expansion into diverse environments. NEW & NOTEWORTHY Oxidative stress results from environmental challenges that increase in frequency and severity during the Anthropocene. Oxygen radical attack causes lipid peroxidation leading to HNE production. Proteome-wide HNE adduction is highly prevalent in Botryllus schlosseri , a widely distributed, highly invasive, and economically important biofouling ascidian and the first marine species to be analyzed for proteome HNE modification. HNE adduction of specific proteins physiologically sequesters reactive oxygen species, which enhances fitness and resilience during environmental change.
Collapse
|
19
|
Zhou C, Zhong Y, Chu Y, Chen R, Wang Y, Zheng Y, Dai H, Zhan C, Xie A, Luo J. Glutathione S-Transferase α4 Alleviates Hyperlipidemia-Induced Vascular Neointimal Hyperplasia in Arteriovenous Grafts via Inhibiting Endoplasmic Reticulum Stress. J Cardiovasc Pharmacol 2024; 84:58-70. [PMID: 38573593 DOI: 10.1097/fjc.0000000000001570] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Accepted: 03/09/2024] [Indexed: 04/05/2024]
Abstract
ABSTRACT Neointimal hyperplasia causes the failure of coronary artery bypass grafting. Our previous studies have found that endothelial dysfunction is 1 candidate for triggering neointimal hyperplasia, but which factors are involved in this process is unclear. Glutathione S-transferase α4 (GSTA4) plays an important role in metabolizing 4-hydroxynonenal (4-HNE), a highly reactive lipid peroxidation product, which causes endothelial dysfunction or death. Here, we investigated the role of GSTA4 in neointima formation after arteriovenous grafts (AVGs) with or without high-fat diet (HFD). Compared with normal diet, HFD caused endothelial dysfunction and increased neointima formation, concomitantly accompanied by downregulated expression of GSTA4 at the mRNA and protein levels. In vitro, overexpression of GSTA4 attenuated 4-HNE-induced endothelial dysfunction and knockdown of GSTA4 aggravated endothelial dysfunction. Furthermore, silencing GSTA4 expression facilitated the activation of 4-HNE-induced endoplasmic reticulum stress and inhibition of endoplasmic reticulum stress pathway alleviated 4-HNE-induced endothelial dysfunction. In addition, compared with wild-type mice, mice with knockout of endothelial-specific GSTA4 (GSTA4 endothelial cell KO) exhibited exacerbated vascular endothelial dysfunction and increased neointima formation caused by HFD. Together, these results demonstrate the critical role of GSTA4 in protecting the function of endothelial cells and in alleviating hyperlipidemia-induced vascular neointimal hyperplasia in arteriovenous grafts.
Collapse
Affiliation(s)
- Chenchen Zhou
- Department of Emergency Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Department of Critical Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yanxia Zhong
- Intensive Care Unit, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China ; and
| | - Yun Chu
- Department of Emergency Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Department of Critical Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Renyu Chen
- Department of Emergency Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Department of Critical Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yurou Wang
- Department of Emergency Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Department of Critical Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yingfang Zheng
- Department of Emergency Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Department of Critical Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hongkai Dai
- Department of Emergency Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Department of Critical Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Chengye Zhan
- Department of Emergency Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Department of Critical Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Aini Xie
- Division of Cardiothoracic and Vascular Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jinlong Luo
- Department of Emergency Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Department of Critical Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
20
|
Aleksic M, Meng X. Protein Haptenation and Its Role in Allergy. Chem Res Toxicol 2024; 37:850-872. [PMID: 38834188 PMCID: PMC11187640 DOI: 10.1021/acs.chemrestox.4c00062] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Revised: 05/14/2024] [Accepted: 05/21/2024] [Indexed: 06/06/2024]
Abstract
Humans are exposed to numerous electrophilic chemicals either as medicines, in the workplace, in nature, or through use of many common cosmetic and household products. Covalent modification of human proteins by such chemicals, or protein haptenation, is a common occurrence in cells and may result in generation of antigenic species, leading to development of hypersensitivity reactions. Ranging in severity of symptoms from local cutaneous reactions and rhinitis to potentially life-threatening anaphylaxis and severe hypersensitivity reactions such as Stephen-Johnson syndrome (SJS) and toxic epidermal necrolysis (TEN), all these reactions have the same Molecular Initiating Event (MIE), i.e. haptenation. However, not all individuals who are exposed to electrophilic chemicals develop symptoms of hypersensitivity. In the present review, we examine common chemistry behind the haptenation reactions leading to formation of neoantigens. We explore simple reactions involving single molecule additions to a nucleophilic side chain of proteins and complex reactions involving multiple electrophilic centers on a single molecule or involving more than one electrophilic molecule as well as the generation of reactive molecules from the interaction with cellular detoxification mechanisms. Besides generation of antigenic species and enabling activation of the immune system, we explore additional events which result directly from the presence of electrophilic chemicals in cells, including activation of key defense mechanisms and immediate consequences of those reactions, and explore their potential effects. We discuss the factors that work in concert with haptenation leading to the development of hypersensitivity reactions and those that may act to prevent it from developing. We also review the potential harnessing of the specificity of haptenation in the design of potent covalent therapeutic inhibitors.
Collapse
Affiliation(s)
- Maja Aleksic
- Safety
and Environmental Assurance Centre, Unilever,
Colworth Science Park, Sharnbrook, Bedford MK44
1LQ, U.K.
| | - Xiaoli Meng
- MRC
Centre for Drug Safety Science, Department of Molecular and Clinical
Pharmacology, The University of Liverpool, Liverpool L69 3GE, U.K.
| |
Collapse
|
21
|
Gupta RC, Singh-Gupta V, Szekely KJ, Zhang K, Lanfear DE, Sabbah HN. Dysregulation of cardiac mitochondrial aldehyde dehydrogenase 2: Studies in dogs with chronic heart failure. JOURNAL OF MOLECULAR AND CELLULAR CARDIOLOGY PLUS 2024; 8:100067. [PMID: 38938550 PMCID: PMC11210280 DOI: 10.1016/j.jmccpl.2024.100067] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 06/29/2024]
Abstract
Mitochondrial (MITO) dysfunction occurs in the failing heart and contributes to worsening of heart failure (HF). Reduced aldehyde dehydrogenase 2 (ALDH2) in left ventricular (LV) myocardium of diabetic hearts has been implicated in MITO dysfunction through accumulation of toxic aldehydes including and elevated levels of 4-hydroxy-2-nonenal (4HNE). This study examined whether dysregulation of MITO ALDH2 (mALDH2) occurs in mitochondria of the failing LV and is associated with increased levels of 4HNE. LV tissue from 7 HF and 7 normal (NL) dogs was obtained. Protein quantification of total mitochondrial ALDH2 (t-mALDH2), phosphorylated mALDH2 (p-mALDH2), total MITO protein kinase c epsilon (t-mPKCε), phosphorylated mPKCε (p-mPKCε) was performed by Western blotting, and total mALDH2 enzymatic activity was measured. Protein adducts of 4HNE-MITO and 4HNE-mALDH2 were also measured in MITO fraction by Western Blotting. Protein level of t-mALDH2 was decreased in HF compared with NL dogs (0.63 ± 0.07 vs 1.17 ± 0.08, p < 0.05) as did mALDH2 enzymatic activity (51.39 ± 3 vs. 107.66 ± 4 nmol NADH/min/mg, p < 0.05). Phosphorylated-mALDH2 and p-mPKCε were unchanged. 4HNE-MITO proteins adduct levels increased in HF compared with NL (2.45 ± 0.08 vs 1.30 ± 0.03 du, p < 0.05) as did adduct levels of 4HNE-mALDH2 (1.60 ± 0.20 vs 0.39 ± 0.08, p < 0.05). In isolated failing cardiomyocytes (CM) exposure to 4HNE decreased mALDH2 activity, increased ROS and 4HNE-ALDH2 adducts, and worsened MITO function. Stimulation of mALDH2 activity with ALDA-1 in isolated HF CMs compared to NL CMs improved ADP-stimulated respiration and maximal ATP synthesis to a greater extant (+47 % and +89 %, respectively). Down-regulation of mALDH2 protein levels and activity occurs in HF and contributes to MITO dysfunction and is likely caused by accumulation of 4HNE-mALDH2 adduct. Increasing mALDH2 activity (via ALDA-1) improved MITO function in failing CMs.
Collapse
Affiliation(s)
- Ramesh C. Gupta
- Department of Medicine, Division of Cardiovascular Medicine, Henry Ford Hospital, 2799 West Grand Boulevard, Detroit, MI 48202, USA
- Henry Ford Health & Michigan State University Health Science, USA
| | - Vinita Singh-Gupta
- Department of Medicine, Division of Cardiovascular Medicine, Henry Ford Hospital, 2799 West Grand Boulevard, Detroit, MI 48202, USA
- Henry Ford Health & Michigan State University Health Science, USA
| | - Kristina J. Szekely
- Department of Medicine, Division of Cardiovascular Medicine, Henry Ford Hospital, 2799 West Grand Boulevard, Detroit, MI 48202, USA
- Henry Ford Health & Michigan State University Health Science, USA
| | - Kefei Zhang
- Department of Medicine, Division of Cardiovascular Medicine, Henry Ford Hospital, 2799 West Grand Boulevard, Detroit, MI 48202, USA
- Henry Ford Health & Michigan State University Health Science, USA
| | - David E. Lanfear
- Department of Medicine, Division of Cardiovascular Medicine, Henry Ford Hospital, 2799 West Grand Boulevard, Detroit, MI 48202, USA
- Henry Ford Health & Michigan State University Health Science, USA
| | - Hani N. Sabbah
- Corresponding author at: Department of Medicine, Division of Cardiovascular Medicine, Henry Ford Hospital, 2799 West Grand Boulevard, Detroit, MI 48202, USA. (H.N. Sabbah)
| |
Collapse
|
22
|
Jung T, Findik N, Hartmann B, Hanack K, Grossmann K, Roggenbuck D, Wegmann M, Mantke R, Deckert M, Grune T. Automated determination of 8-OHdG in cells and tissue via immunofluorescence using a specially created antibody. BIOTECHNOLOGY REPORTS (AMSTERDAM, NETHERLANDS) 2024; 42:e00833. [PMID: 38948353 PMCID: PMC11211097 DOI: 10.1016/j.btre.2024.e00833] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Revised: 02/27/2024] [Accepted: 02/27/2024] [Indexed: 07/02/2024]
Abstract
Despite powerful DNA repair systems, oxidative damage/modification to DNA is an inevitable side effect of metabolism, ionizing radiation, lifestyle habits, inflammatory pathologies such as type-2 diabetes or metabolic syndrome, cancer and natural aging. One of the most common oxidative DNA modifications is 8-OHdG (8‑hydroxy-2'-deoxyguanosine), which is the most widely used marker in research and clinical diagnostics. 8-OHdG is easily and specifically detectable in various samples such as urine, plasma, cells and tissues via a large variety of methods like ELISA, HPLC, chromatographic methods, and immunochemistry. Formed by oxidation of guanine and being representative for the degree of DNA damage, 8-OHdG can be also used as biomarker for risk assessment of various cancers as well as degenerative diseases. Here, we present a highly specific, self-developed 8-OHdG antibody in successful comparison to a commercially one, tested in cells (FF95, HCT116, and HT22) and intestinal tissue, focusing on automatized evaluation via fluorescence/confocal microscopy.
Collapse
Affiliation(s)
- Tobias Jung
- Department of Molecular Toxicology, German Institute of Human Nutrition Potsdam-Rehbruecke (DIfE), Arthur-Scheunert-Allee 114-116 14558, Nuthetal, Germany
- German Center for Cardiovascular Research (DZHK), 10117, Berlin, Germany
| | - Nicole Findik
- new/era/mabs GmbH, August-Bebel-Str. 89 14482 Potsdam, Germany
| | - Bianca Hartmann
- new/era/mabs GmbH, August-Bebel-Str. 89 14482 Potsdam, Germany
| | - Katja Hanack
- new/era/mabs GmbH, August-Bebel-Str. 89 14482 Potsdam, Germany
- University of Potsdam, Department of Biochemistry and Biology, Chair of Immunotechnology, Karl-Liebknecht-Str. 24-25, Build 29, Office 1.55 14476 Potsdam, Germany
| | | | - Dirk Roggenbuck
- Faculty of Health Sciences Brandenburg, Brandenburg Technical University Cottbus-Senftenberg
| | - Marc Wegmann
- MEDIPAN GmbH, Ludwig-Erhard-Ring 3 15827 Dahlewitz
| | - René Mantke
- Brandenburg Medical School Theodor Fontane, Klinik für Allgemein- und Viszeralchirurgie, Klinikum Brandenburg, Hochstraße 29 14770 Brandenburg an der Havel, Germany
| | - Markus Deckert
- Brandenburg Medical School Theodor Fontane, Theodor Fontane Campus Brandenburg, Hämatologie, Onkologie SKB, IAG Psychoonkologie und Palliativversorgung, Hochstraße 29, 14770 Brandenburg an der Havel, Germany
| | - Tilman Grune
- Department of Molecular Toxicology, German Institute of Human Nutrition Potsdam-Rehbruecke (DIfE), Arthur-Scheunert-Allee 114-116 14558, Nuthetal, Germany
- German Center for Cardiovascular Research (DZHK), 10117, Berlin, Germany
- German Center for Diabetes Research (DZD) 85764 Muenchen-Neuherberg, Germany
- NutriAct – Competence Cluster Nutrition Research Berlin-Potsdam, 14558 Nuthetal, Germany
- University of Potsdam, Institute of Nutrition 14558 Nuthetal, Germany
| |
Collapse
|
23
|
Thomas KM, Spitzer N. Silver nanoparticles induce formation of multi-protein aggregates that contain cadherin but do not colocalize with nanoparticles. Toxicol In Vitro 2024; 98:105837. [PMID: 38692336 DOI: 10.1016/j.tiv.2024.105837] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Revised: 04/24/2024] [Accepted: 04/28/2024] [Indexed: 05/03/2024]
Abstract
Silver nanoparticles (AgNPs) are increasingly incorporated in diverse products to confer antimicrobial properties. They are released into the environment during manufacture, after disposal, and from the products during use. Because AgNPs bioaccumulate in brain, it is important to understand how they interact with neural cell physiology. We found that the focal adhesion (FA)-associated protein cadherin aggregated in a dose-dependent response to AgNP exposure in differentiating cultured B35 neuroblastoma cells. These aggregates tended to colocalize with F-actin inclusions that form in response to AgNP and also contain β-catenin. However, using hyperspectral microscopy, we demonstrate that these multi-protein aggregates did not colocalize with the AgNPs themselves. Furthermore, expression and organization of the FA protein vinculin did not change in cells exposed to AgNP. Our findings suggest that AgNPs activate an intermediate mechanism which leads to formation of aggregates via specific protein-protein interactions. Finally, we detail the changes in hyperspectral profiles of AgNPs during different stages of cell culture and immunocytochemistry processing. AgNPs in citrate-stabilized solution present mostly blue with some rainbow spectra and these are maintained upon mounting in Prolong Gold. Exposure to tissue culture medium results in a uniform green spectral shift that is not further altered by fixation and protein block steps of immunocytochemistry.
Collapse
Affiliation(s)
- Kaden M Thomas
- Department of Biological Sciences, Marshall University, One John Marshall Dr., Huntington, WV, USA
| | - Nadja Spitzer
- Department of Biological Sciences, Marshall University, One John Marshall Dr., Huntington, WV, USA.
| |
Collapse
|
24
|
Zhai X, Cao S, Wang J, Qiao B, Liu X, Hua R, Zhao M, Sun S, Han Y, Wu S, Pang J, Yuan Q, Wang B, Xu F, Wei S, Chen Y. Carbonylation of Runx2 at K176 by 4-Hydroxynonenal Accelerates Vascular Calcification. Circulation 2024; 149:1752-1769. [PMID: 38348663 DOI: 10.1161/circulationaha.123.065830] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/04/2023] [Accepted: 01/19/2024] [Indexed: 05/30/2024]
Abstract
BACKGROUND Vascular calcification, which is characterized by calcium deposition in arterial walls and the osteochondrogenic differentiation of vascular smooth muscle cells, is an actively regulated process that involves complex mechanisms. Vascular calcification is associated with increased cardiovascular adverse events. The role of 4-hydroxynonenal (4-HNE), which is the most abundant stable product of lipid peroxidation, in vascular calcification has been poorly investigated. METHODS Serum was collected from patients with chronic kidney disease and controls, and the levels of 4-HNE and 8-iso-prostaglandin F2α were measured. Sections of coronary atherosclerotic plaques from donors were immunostained to analyze calcium deposition and 4-HNE. A total of 658 patients with coronary artery disease who received coronary computed tomography angiography were recruited to analyze the relationship between coronary calcification and the rs671 mutation in aldehyde dehydrogenase 2 (ALDH2). ALDH2 knockout (ALDH2-/-) mice, smooth muscle cell-specific ALDH2 knockout mice, ALDH2 transgenic mice, and their controls were used to establish vascular calcification models. Primary mouse aortic smooth muscle cells and human aortic smooth muscle cells were exposed to medium containing β-glycerophosphate and CaCl2 to investigate cell calcification and the underlying molecular mechanisms. RESULTS Elevated 4-HNE levels were observed in the serum of patients with chronic kidney disease and model mice and were detected in calcified artery sections by immunostaining. ALDH2 knockout or smooth muscle cell-specific ALDH2 knockout accelerated the development of vascular calcification in model mice, whereas overexpression or activation prevented mouse vascular calcification and the osteochondrogenic differentiation of vascular smooth muscle cells. In patients with coronary artery disease, patients with ALDH2 rs671 gene mutation developed more severe coronary calcification. 4-HNE promoted calcification of both mouse aortic smooth muscle cells and human aortic smooth muscle cells and their osteochondrogenic differentiation in vitro. 4-HNE increased the level of Runx2 (runt-related transcription factor-2), and the effect of 4-HNE on promoting vascular smooth muscle cell calcification was ablated when Runx2 was knocked down. Mutation of Runx2 at lysine 176 reduced its carbonylation and eliminated the 4-HNE-induced upregulation of Runx2. CONCLUSIONS Our results suggest that 4-HNE increases Runx2 stabilization by directly carbonylating its K176 site and promotes vascular calcification. ALDH2 might be a potential target for the treatment of vascular calcification.
Collapse
MESH Headings
- Animals
- Aldehydes/metabolism
- Vascular Calcification/metabolism
- Vascular Calcification/genetics
- Vascular Calcification/pathology
- Humans
- Core Binding Factor Alpha 1 Subunit/metabolism
- Core Binding Factor Alpha 1 Subunit/genetics
- Aldehyde Dehydrogenase, Mitochondrial/genetics
- Aldehyde Dehydrogenase, Mitochondrial/metabolism
- Mice
- Mice, Knockout
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/pathology
- Myocytes, Smooth Muscle/drug effects
- Male
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/pathology
- Female
- Middle Aged
- Coronary Artery Disease/metabolism
- Coronary Artery Disease/genetics
- Coronary Artery Disease/pathology
- Cells, Cultured
- Renal Insufficiency, Chronic/metabolism
- Renal Insufficiency, Chronic/genetics
- Renal Insufficiency, Chronic/pathology
- Aged
Collapse
Affiliation(s)
- Xiaoxuan Zhai
- Department of Emergency and Chest Pain Center, Qilu Hospital of Shandong University, Jinan, China (X.Z., S.C., J.W., B.Q., X.L., R.H., M.Z., S.S., Y.H., S.W., J.P., Q.Y., F.X., S.W., Y.C.)
- Shandong Provincial Clinical Research Center for Emergency and Critical Care Medicine, Institute of Emergency and Critical Care Medicine of Shandong University, Qilu Hospital of Shandong University, Jinan, China (X.Z., S.C., J.W., B.Q., X.L., R.H., M.Z., S.S., Y.H., S.W., J.P., Q.Y., F.X., S.W., Y.C.)
- Key Laboratory of Emergency and Critical Care Medicine of Shandong Province, Key Laboratory of Cardiopulmonary-Cerebral Resuscitation Research of Shandong Province, Qilu Hospital of Shandong University, Jinan, China (X.Z., S.C., J.W., B.Q., X.L., R.H., M.Z., S.S., Y.H., S.W., J.P., Q.Y., F.X., S.W., Y.C.)
| | - Shengchuan Cao
- Department of Emergency and Chest Pain Center, Qilu Hospital of Shandong University, Jinan, China (X.Z., S.C., J.W., B.Q., X.L., R.H., M.Z., S.S., Y.H., S.W., J.P., Q.Y., F.X., S.W., Y.C.)
- Shandong Provincial Clinical Research Center for Emergency and Critical Care Medicine, Institute of Emergency and Critical Care Medicine of Shandong University, Qilu Hospital of Shandong University, Jinan, China (X.Z., S.C., J.W., B.Q., X.L., R.H., M.Z., S.S., Y.H., S.W., J.P., Q.Y., F.X., S.W., Y.C.)
- Key Laboratory of Emergency and Critical Care Medicine of Shandong Province, Key Laboratory of Cardiopulmonary-Cerebral Resuscitation Research of Shandong Province, Qilu Hospital of Shandong University, Jinan, China (X.Z., S.C., J.W., B.Q., X.L., R.H., M.Z., S.S., Y.H., S.W., J.P., Q.Y., F.X., S.W., Y.C.)
| | - Jiali Wang
- Department of Emergency and Chest Pain Center, Qilu Hospital of Shandong University, Jinan, China (X.Z., S.C., J.W., B.Q., X.L., R.H., M.Z., S.S., Y.H., S.W., J.P., Q.Y., F.X., S.W., Y.C.)
- Shandong Provincial Clinical Research Center for Emergency and Critical Care Medicine, Institute of Emergency and Critical Care Medicine of Shandong University, Qilu Hospital of Shandong University, Jinan, China (X.Z., S.C., J.W., B.Q., X.L., R.H., M.Z., S.S., Y.H., S.W., J.P., Q.Y., F.X., S.W., Y.C.)
- Key Laboratory of Emergency and Critical Care Medicine of Shandong Province, Key Laboratory of Cardiopulmonary-Cerebral Resuscitation Research of Shandong Province, Qilu Hospital of Shandong University, Jinan, China (X.Z., S.C., J.W., B.Q., X.L., R.H., M.Z., S.S., Y.H., S.W., J.P., Q.Y., F.X., S.W., Y.C.)
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese Ministry of Health and Chinese Academy of Medical Sciences, Qilu Hospital of Shandong University, Jinan (J.W., B.Q., X.L., R.H., M.Z., S.S., Y.H., J.P., Q.Y.)
| | - Bao Qiao
- Department of Emergency and Chest Pain Center, Qilu Hospital of Shandong University, Jinan, China (X.Z., S.C., J.W., B.Q., X.L., R.H., M.Z., S.S., Y.H., S.W., J.P., Q.Y., F.X., S.W., Y.C.)
- Shandong Provincial Clinical Research Center for Emergency and Critical Care Medicine, Institute of Emergency and Critical Care Medicine of Shandong University, Qilu Hospital of Shandong University, Jinan, China (X.Z., S.C., J.W., B.Q., X.L., R.H., M.Z., S.S., Y.H., S.W., J.P., Q.Y., F.X., S.W., Y.C.)
- Key Laboratory of Emergency and Critical Care Medicine of Shandong Province, Key Laboratory of Cardiopulmonary-Cerebral Resuscitation Research of Shandong Province, Qilu Hospital of Shandong University, Jinan, China (X.Z., S.C., J.W., B.Q., X.L., R.H., M.Z., S.S., Y.H., S.W., J.P., Q.Y., F.X., S.W., Y.C.)
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese Ministry of Health and Chinese Academy of Medical Sciences, Qilu Hospital of Shandong University, Jinan (J.W., B.Q., X.L., R.H., M.Z., S.S., Y.H., J.P., Q.Y.)
| | - Xuehao Liu
- Department of Emergency and Chest Pain Center, Qilu Hospital of Shandong University, Jinan, China (X.Z., S.C., J.W., B.Q., X.L., R.H., M.Z., S.S., Y.H., S.W., J.P., Q.Y., F.X., S.W., Y.C.)
- Shandong Provincial Clinical Research Center for Emergency and Critical Care Medicine, Institute of Emergency and Critical Care Medicine of Shandong University, Qilu Hospital of Shandong University, Jinan, China (X.Z., S.C., J.W., B.Q., X.L., R.H., M.Z., S.S., Y.H., S.W., J.P., Q.Y., F.X., S.W., Y.C.)
- Key Laboratory of Emergency and Critical Care Medicine of Shandong Province, Key Laboratory of Cardiopulmonary-Cerebral Resuscitation Research of Shandong Province, Qilu Hospital of Shandong University, Jinan, China (X.Z., S.C., J.W., B.Q., X.L., R.H., M.Z., S.S., Y.H., S.W., J.P., Q.Y., F.X., S.W., Y.C.)
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese Ministry of Health and Chinese Academy of Medical Sciences, Qilu Hospital of Shandong University, Jinan (J.W., B.Q., X.L., R.H., M.Z., S.S., Y.H., J.P., Q.Y.)
| | - Rui Hua
- Department of Emergency and Chest Pain Center, Qilu Hospital of Shandong University, Jinan, China (X.Z., S.C., J.W., B.Q., X.L., R.H., M.Z., S.S., Y.H., S.W., J.P., Q.Y., F.X., S.W., Y.C.)
- Shandong Provincial Clinical Research Center for Emergency and Critical Care Medicine, Institute of Emergency and Critical Care Medicine of Shandong University, Qilu Hospital of Shandong University, Jinan, China (X.Z., S.C., J.W., B.Q., X.L., R.H., M.Z., S.S., Y.H., S.W., J.P., Q.Y., F.X., S.W., Y.C.)
- Key Laboratory of Emergency and Critical Care Medicine of Shandong Province, Key Laboratory of Cardiopulmonary-Cerebral Resuscitation Research of Shandong Province, Qilu Hospital of Shandong University, Jinan, China (X.Z., S.C., J.W., B.Q., X.L., R.H., M.Z., S.S., Y.H., S.W., J.P., Q.Y., F.X., S.W., Y.C.)
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese Ministry of Health and Chinese Academy of Medical Sciences, Qilu Hospital of Shandong University, Jinan (J.W., B.Q., X.L., R.H., M.Z., S.S., Y.H., J.P., Q.Y.)
| | - Menglin Zhao
- Department of Emergency and Chest Pain Center, Qilu Hospital of Shandong University, Jinan, China (X.Z., S.C., J.W., B.Q., X.L., R.H., M.Z., S.S., Y.H., S.W., J.P., Q.Y., F.X., S.W., Y.C.)
- Shandong Provincial Clinical Research Center for Emergency and Critical Care Medicine, Institute of Emergency and Critical Care Medicine of Shandong University, Qilu Hospital of Shandong University, Jinan, China (X.Z., S.C., J.W., B.Q., X.L., R.H., M.Z., S.S., Y.H., S.W., J.P., Q.Y., F.X., S.W., Y.C.)
- Key Laboratory of Emergency and Critical Care Medicine of Shandong Province, Key Laboratory of Cardiopulmonary-Cerebral Resuscitation Research of Shandong Province, Qilu Hospital of Shandong University, Jinan, China (X.Z., S.C., J.W., B.Q., X.L., R.H., M.Z., S.S., Y.H., S.W., J.P., Q.Y., F.X., S.W., Y.C.)
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese Ministry of Health and Chinese Academy of Medical Sciences, Qilu Hospital of Shandong University, Jinan (J.W., B.Q., X.L., R.H., M.Z., S.S., Y.H., J.P., Q.Y.)
| | - Shukun Sun
- Department of Emergency and Chest Pain Center, Qilu Hospital of Shandong University, Jinan, China (X.Z., S.C., J.W., B.Q., X.L., R.H., M.Z., S.S., Y.H., S.W., J.P., Q.Y., F.X., S.W., Y.C.)
- Shandong Provincial Clinical Research Center for Emergency and Critical Care Medicine, Institute of Emergency and Critical Care Medicine of Shandong University, Qilu Hospital of Shandong University, Jinan, China (X.Z., S.C., J.W., B.Q., X.L., R.H., M.Z., S.S., Y.H., S.W., J.P., Q.Y., F.X., S.W., Y.C.)
- Key Laboratory of Emergency and Critical Care Medicine of Shandong Province, Key Laboratory of Cardiopulmonary-Cerebral Resuscitation Research of Shandong Province, Qilu Hospital of Shandong University, Jinan, China (X.Z., S.C., J.W., B.Q., X.L., R.H., M.Z., S.S., Y.H., S.W., J.P., Q.Y., F.X., S.W., Y.C.)
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese Ministry of Health and Chinese Academy of Medical Sciences, Qilu Hospital of Shandong University, Jinan (J.W., B.Q., X.L., R.H., M.Z., S.S., Y.H., J.P., Q.Y.)
| | - Yu Han
- Department of Emergency and Chest Pain Center, Qilu Hospital of Shandong University, Jinan, China (X.Z., S.C., J.W., B.Q., X.L., R.H., M.Z., S.S., Y.H., S.W., J.P., Q.Y., F.X., S.W., Y.C.)
- Shandong Provincial Clinical Research Center for Emergency and Critical Care Medicine, Institute of Emergency and Critical Care Medicine of Shandong University, Qilu Hospital of Shandong University, Jinan, China (X.Z., S.C., J.W., B.Q., X.L., R.H., M.Z., S.S., Y.H., S.W., J.P., Q.Y., F.X., S.W., Y.C.)
- Key Laboratory of Emergency and Critical Care Medicine of Shandong Province, Key Laboratory of Cardiopulmonary-Cerebral Resuscitation Research of Shandong Province, Qilu Hospital of Shandong University, Jinan, China (X.Z., S.C., J.W., B.Q., X.L., R.H., M.Z., S.S., Y.H., S.W., J.P., Q.Y., F.X., S.W., Y.C.)
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese Ministry of Health and Chinese Academy of Medical Sciences, Qilu Hospital of Shandong University, Jinan (J.W., B.Q., X.L., R.H., M.Z., S.S., Y.H., J.P., Q.Y.)
| | - Shuo Wu
- Department of Emergency and Chest Pain Center, Qilu Hospital of Shandong University, Jinan, China (X.Z., S.C., J.W., B.Q., X.L., R.H., M.Z., S.S., Y.H., S.W., J.P., Q.Y., F.X., S.W., Y.C.)
- Shandong Provincial Clinical Research Center for Emergency and Critical Care Medicine, Institute of Emergency and Critical Care Medicine of Shandong University, Qilu Hospital of Shandong University, Jinan, China (X.Z., S.C., J.W., B.Q., X.L., R.H., M.Z., S.S., Y.H., S.W., J.P., Q.Y., F.X., S.W., Y.C.)
- Key Laboratory of Emergency and Critical Care Medicine of Shandong Province, Key Laboratory of Cardiopulmonary-Cerebral Resuscitation Research of Shandong Province, Qilu Hospital of Shandong University, Jinan, China (X.Z., S.C., J.W., B.Q., X.L., R.H., M.Z., S.S., Y.H., S.W., J.P., Q.Y., F.X., S.W., Y.C.)
| | - Jiaojiao Pang
- Department of Emergency and Chest Pain Center, Qilu Hospital of Shandong University, Jinan, China (X.Z., S.C., J.W., B.Q., X.L., R.H., M.Z., S.S., Y.H., S.W., J.P., Q.Y., F.X., S.W., Y.C.)
- Shandong Provincial Clinical Research Center for Emergency and Critical Care Medicine, Institute of Emergency and Critical Care Medicine of Shandong University, Qilu Hospital of Shandong University, Jinan, China (X.Z., S.C., J.W., B.Q., X.L., R.H., M.Z., S.S., Y.H., S.W., J.P., Q.Y., F.X., S.W., Y.C.)
- Key Laboratory of Emergency and Critical Care Medicine of Shandong Province, Key Laboratory of Cardiopulmonary-Cerebral Resuscitation Research of Shandong Province, Qilu Hospital of Shandong University, Jinan, China (X.Z., S.C., J.W., B.Q., X.L., R.H., M.Z., S.S., Y.H., S.W., J.P., Q.Y., F.X., S.W., Y.C.)
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese Ministry of Health and Chinese Academy of Medical Sciences, Qilu Hospital of Shandong University, Jinan (J.W., B.Q., X.L., R.H., M.Z., S.S., Y.H., J.P., Q.Y.)
| | - Qiuhuan Yuan
- Department of Emergency and Chest Pain Center, Qilu Hospital of Shandong University, Jinan, China (X.Z., S.C., J.W., B.Q., X.L., R.H., M.Z., S.S., Y.H., S.W., J.P., Q.Y., F.X., S.W., Y.C.)
- Shandong Provincial Clinical Research Center for Emergency and Critical Care Medicine, Institute of Emergency and Critical Care Medicine of Shandong University, Qilu Hospital of Shandong University, Jinan, China (X.Z., S.C., J.W., B.Q., X.L., R.H., M.Z., S.S., Y.H., S.W., J.P., Q.Y., F.X., S.W., Y.C.)
- Key Laboratory of Emergency and Critical Care Medicine of Shandong Province, Key Laboratory of Cardiopulmonary-Cerebral Resuscitation Research of Shandong Province, Qilu Hospital of Shandong University, Jinan, China (X.Z., S.C., J.W., B.Q., X.L., R.H., M.Z., S.S., Y.H., S.W., J.P., Q.Y., F.X., S.W., Y.C.)
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese Ministry of Health and Chinese Academy of Medical Sciences, Qilu Hospital of Shandong University, Jinan (J.W., B.Q., X.L., R.H., M.Z., S.S., Y.H., J.P., Q.Y.)
| | - Bailu Wang
- National Medical Products Administration Key Laboratory for Clinical Research and Evaluation of Innovative Drug, Clinical Trial Center, Qilu Hospital of Shandong University, Jinan, China (B.W.)
| | - Feng Xu
- Department of Emergency and Chest Pain Center, Qilu Hospital of Shandong University, Jinan, China (X.Z., S.C., J.W., B.Q., X.L., R.H., M.Z., S.S., Y.H., S.W., J.P., Q.Y., F.X., S.W., Y.C.)
- Shandong Provincial Clinical Research Center for Emergency and Critical Care Medicine, Institute of Emergency and Critical Care Medicine of Shandong University, Qilu Hospital of Shandong University, Jinan, China (X.Z., S.C., J.W., B.Q., X.L., R.H., M.Z., S.S., Y.H., S.W., J.P., Q.Y., F.X., S.W., Y.C.)
- Key Laboratory of Emergency and Critical Care Medicine of Shandong Province, Key Laboratory of Cardiopulmonary-Cerebral Resuscitation Research of Shandong Province, Qilu Hospital of Shandong University, Jinan, China (X.Z., S.C., J.W., B.Q., X.L., R.H., M.Z., S.S., Y.H., S.W., J.P., Q.Y., F.X., S.W., Y.C.)
| | - Shujian Wei
- Department of Emergency and Chest Pain Center, Qilu Hospital of Shandong University, Jinan, China (X.Z., S.C., J.W., B.Q., X.L., R.H., M.Z., S.S., Y.H., S.W., J.P., Q.Y., F.X., S.W., Y.C.)
- Shandong Provincial Clinical Research Center for Emergency and Critical Care Medicine, Institute of Emergency and Critical Care Medicine of Shandong University, Qilu Hospital of Shandong University, Jinan, China (X.Z., S.C., J.W., B.Q., X.L., R.H., M.Z., S.S., Y.H., S.W., J.P., Q.Y., F.X., S.W., Y.C.)
- Key Laboratory of Emergency and Critical Care Medicine of Shandong Province, Key Laboratory of Cardiopulmonary-Cerebral Resuscitation Research of Shandong Province, Qilu Hospital of Shandong University, Jinan, China (X.Z., S.C., J.W., B.Q., X.L., R.H., M.Z., S.S., Y.H., S.W., J.P., Q.Y., F.X., S.W., Y.C.)
| | - Yuguo Chen
- Department of Emergency and Chest Pain Center, Qilu Hospital of Shandong University, Jinan, China (X.Z., S.C., J.W., B.Q., X.L., R.H., M.Z., S.S., Y.H., S.W., J.P., Q.Y., F.X., S.W., Y.C.)
- Shandong Provincial Clinical Research Center for Emergency and Critical Care Medicine, Institute of Emergency and Critical Care Medicine of Shandong University, Qilu Hospital of Shandong University, Jinan, China (X.Z., S.C., J.W., B.Q., X.L., R.H., M.Z., S.S., Y.H., S.W., J.P., Q.Y., F.X., S.W., Y.C.)
- Key Laboratory of Emergency and Critical Care Medicine of Shandong Province, Key Laboratory of Cardiopulmonary-Cerebral Resuscitation Research of Shandong Province, Qilu Hospital of Shandong University, Jinan, China (X.Z., S.C., J.W., B.Q., X.L., R.H., M.Z., S.S., Y.H., S.W., J.P., Q.Y., F.X., S.W., Y.C.)
| |
Collapse
|
25
|
Ovchinnikov AN, Paoli A. Saliva as a Diagnostic Tool for Early Detection of Exercise-Induced Oxidative Damage in Female Athletes. Biomedicines 2024; 12:1006. [PMID: 38790968 PMCID: PMC11118847 DOI: 10.3390/biomedicines12051006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2024] [Revised: 04/17/2024] [Accepted: 04/24/2024] [Indexed: 05/26/2024] Open
Abstract
Although blood still remains the most commonly utilized medium to detect increased levels of oxidative damage induced by exercise, saliva diagnostics have gained increasing popularity due to their non-invasive nature and athlete-friendly collection process. Given that the contribution of various phases of the menstrual cycle to the levels of oxidative damage may differ, the aim of this study was to evaluate an agreement between salivary and plasmatic levels of lipid peroxidation products in female swimmers in both the follicular (F) and luteal (L) phases of the menstrual cycle at rest and following exercise. Twelve well-trained female swimmers aged 19.6 ± 1.1 years old were examined. We measured diene conjugates (DCs), triene conjugates (TCs), and Schiff bases (SBs) in lipids immediately after their extraction from both saliva and blood plasma. All female swimmers were studied two times each, in the two different phases of one menstrual cycle, before and after high-intensity interval exercise (HIIE). Salivary and plasmatic levels of DCs, TCs, and SBs significantly increased post-exercise compared to pre-exercise, in both the F and L phases. A high positive correlation was observed between the concentrations of DCs, TCs, and SBs in the saliva and blood plasma of participants in the F and L phases, both at rest and following HIIE. Ordinary least products regression analysis indicates that there was no proportional and differential bias in the data. The Bland-Altman method also declares that there was no differential bias, since the line of equality was within the 95% confidence interval of the mean difference between salivary and plasmatic levels of DCs, TCs, and SBs in female swimmers, in both the F and L phases, before and after HIIE. There was also no proportional bias in the Bland-Altman plots. Thus, this is the first study to report a high agreement between the quantifications of DCs, TCs, and SBs in the saliva and blood plasma of female swimmers in both the F and L phases, at rest and following HIIE.
Collapse
Affiliation(s)
- Aleksandr N. Ovchinnikov
- Laboratory of Non-Invasive Diagnostics in Sports, Department of Sports Medicine and Psychology, Lobachevsky University, 603022 Nizhny Novgorod, Russia
| | - Antonio Paoli
- Department of Biomedical Sciences, University of Padua, 35122 Padua, Italy;
| |
Collapse
|
26
|
Tufail N, Abidi M, Warsi MS, Kausar T, Nayeem SM. Computational and physicochemical insight into 4-hydroxy-2-nonenal induced structural and functional perturbations in human low-density lipoprotein. J Biomol Struct Dyn 2024; 42:2698-2713. [PMID: 37154523 DOI: 10.1080/07391102.2023.2208234] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Accepted: 04/19/2023] [Indexed: 05/10/2023]
Abstract
Lipid peroxidation (LPO) is a biological process that frequently occurs under physiological conditions. Undue oxidative stress increases the level of LPO; which may further contribute to the development of cancer. 4-Hydroxy-2-nonenal (HNE), one of the principal by-products of LPO, is present in high concentrations in oxidatively stressed cells. HNE rapidly reacts with various biological components, including DNA and proteins; however, the extent of protein degradation by lipid electrophiles is not well understood. The influence of HNE on protein structures will likely have a considerable therapeutic value. This research elucidates the potential of HNE, one of the most researched phospholipid peroxidation products, in modifying low-density lipoprotein (LDL). In this study, we tracked the structural alterations in LDL by HNE using various physicochemical techniques. To comprehend the stability, binding mechanism and conformational dynamics of the HNE-LDL complex, computational investigations were carried out. LDL was altered in vitro by HNE, and the secondary and tertiary structural alterations were examined using spectroscopic methods, such as UV-visible, fluorescence, circular dichroism and fourier transform infrared spectroscopy. Carbonyl content, thiobarbituric acid-reactive-substance (TBARS) and nitroblue tetrazolium (NBT) reduction assays were used to examine changes in the oxidation status of LDL. Thioflavin T (ThT), 1-anilinonaphthalene-8-sulfonic (ANS) binding assay and electron microscopy were used to investigate aggregates formation. According to our research, LDL modified by HNE results in changes in structural dynamics, oxidative stress and the formation of LDL aggregates. The current investigation must characterize HNE's interactions with LDL and comprehend how it can change their physiological or pathological functions.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Neda Tufail
- Department of Biochemistry, Jawaharlal Nehru Medical College, Aligarh Muslim University, Aligarh, India
| | - Minhal Abidi
- Department of Biochemistry, Jawaharlal Nehru Medical College, Aligarh Muslim University, Aligarh, India
| | - Mohd Sharib Warsi
- Department of Biochemistry, Jawaharlal Nehru Medical College, Aligarh Muslim University, Aligarh, India
| | - Tasneem Kausar
- Department of Chemistry, Aligarh Muslim University, Aligarh, India
| | - Shahid M Nayeem
- Department of Chemistry, Aligarh Muslim University, Aligarh, India
| |
Collapse
|
27
|
Nègre-Salvayre A, Salvayre R. Reactive Carbonyl Species and Protein Lipoxidation in Atherogenesis. Antioxidants (Basel) 2024; 13:232. [PMID: 38397830 PMCID: PMC10886358 DOI: 10.3390/antiox13020232] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2023] [Revised: 02/08/2024] [Accepted: 02/09/2024] [Indexed: 02/25/2024] Open
Abstract
Atherosclerosis is a multifactorial disease of medium and large arteries, characterized by the presence of lipid-rich plaques lining the intima over time. It is the main cause of cardiovascular diseases and death worldwide. Redox imbalance and lipid peroxidation could play key roles in atherosclerosis by promoting a bundle of responses, including endothelial activation, inflammation, and foam cell formation. The oxidation of polyunsaturated fatty acids generates various lipid oxidation products such as reactive carbonyl species (RCS), including 4-hydroxy alkenals, malondialdehyde, and acrolein. RCS covalently bind to nucleophilic groups of nucleic acids, phospholipids, and proteins, modifying their structure and activity and leading to their progressive dysfunction. Protein lipoxidation is the non-enzymatic post-translational modification of proteins by RCS. Low-density lipoprotein (LDL) oxidation and apolipoprotein B (apoB) modification by RCS play a major role in foam cell formation. Moreover, oxidized LDLs are a source of RCS, which form adducts on a huge number of proteins, depending on oxidative stress intensity, the nature of targets, and the availability of detoxifying systems. Many systems are affected by lipoxidation, including extracellular matrix components, membranes, cytoplasmic and cytoskeletal proteins, transcription factors, and other components. The mechanisms involved in lipoxidation-induced vascular dysfunction are not fully elucidated. In this review, we focus on protein lipoxidation during atherogenesis.
Collapse
Affiliation(s)
- Anne Nègre-Salvayre
- Inserm Unité Mixte de Recherche (UMR), 1297 Toulouse, Centre Hospitalier Universitaire (CHU) Rangueil—BP 84225, 31432 Toulouse CEDEX 4, France;
- Faculty of Medicine, University of Toulouse, 31432 Toulouse, France
| | - Robert Salvayre
- Inserm Unité Mixte de Recherche (UMR), 1297 Toulouse, Centre Hospitalier Universitaire (CHU) Rangueil—BP 84225, 31432 Toulouse CEDEX 4, France;
- Faculty of Medicine, University of Toulouse, 31432 Toulouse, France
| |
Collapse
|
28
|
Díaz M, Valdés-Baizabal C, de Pablo DP, Marin R. Age-Dependent Changes in Nrf2/Keap1 and Target Antioxidant Protein Expression Correlate to Lipoxidative Adducts, and Are Modulated by Dietary N-3 LCPUFA in the Hippocampus of Mice. Antioxidants (Basel) 2024; 13:206. [PMID: 38397804 PMCID: PMC10886099 DOI: 10.3390/antiox13020206] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2023] [Revised: 01/26/2024] [Accepted: 01/30/2024] [Indexed: 02/25/2024] Open
Abstract
The brain has a high metabolism rate that may generate reactive oxygen and nitrogen species. Consequently, nerve cells require highly efficient antioxidant defenses in order to prevent a condition of deleterious oxidative stress. This is particularly relevant in the hippocampus, a highly complex cerebral area involved in processing superior cognitive functions. Most current evidence points to hippocampal oxidative damage as a causal effect for neurodegenerative disorders, especially Alzheimer's disease. Nuclear factor erythroid-2-related factor 2/Kelch-like ECH-associated protein 1 (Nrf2/Keap1) is a master key for the transcriptional regulation of antioxidant and detoxifying systems. It is ubiquitously expressed in brain areas, mainly supporting glial cells. In the present study, we have analyzed the relationships between Nrf2 and Keap1 isoforms in hippocampal tissue in response to aging and dietary long-chain polyunsaturated fatty acids (LCPUFA) supplementation. The possible involvement of lipoxidative and nitrosative by-products in the dynamics of the Nrf2/Keap1 complex was examined though determination of protein adducts, namely malondialdehyde (MDA), 4-hydroxynonenal (HNE), and 3-nitro-tyrosine (NTyr) under basal conditions. The results were correlated to the expression of target proteins heme-oxygenase-1 (HO-1) and glutathione peroxidase 4 (GPx4), whose expressions are known to be regulated by Nrf2/Keap1 signaling activation. All variables in this study were obtained simultaneously from the same preparations, allowing multivariate approaches. The results demonstrate a complex modification of the protein expression patterns together with the formation of adducts in response to aging and diet supplementation. Both parameters exhibited a strong interaction. Noticeably, LCPUFA supplementation to aged animals restored the Nrf2/Keap1/target protein patterns to the status observed in young animals, therefore driving a "rejuvenation" of hippocampal antioxidant defense.
Collapse
Affiliation(s)
- Mario Díaz
- Department of Physics, Faculty of Sciences, University of La Laguna, 38200 Tenerife, Spain
- Instituto Universitario de Neurociencias (IUNE), University of La Laguna, 38320 Tenerife, Spain; (C.V.-B.); (D.P.d.P.); (R.M.)
| | - Catalina Valdés-Baizabal
- Instituto Universitario de Neurociencias (IUNE), University of La Laguna, 38320 Tenerife, Spain; (C.V.-B.); (D.P.d.P.); (R.M.)
- Laboratory of Cellular Neurobiology, Department of Basic Medical Sciences, Faculty of Health Sciences, University of La Laguna, 38200 Tenerife, Spain
| | - Daniel Pereda de Pablo
- Instituto Universitario de Neurociencias (IUNE), University of La Laguna, 38320 Tenerife, Spain; (C.V.-B.); (D.P.d.P.); (R.M.)
- Laboratory of Cellular Neurobiology, Department of Basic Medical Sciences, Faculty of Health Sciences, University of La Laguna, 38200 Tenerife, Spain
| | - Raquel Marin
- Instituto Universitario de Neurociencias (IUNE), University of La Laguna, 38320 Tenerife, Spain; (C.V.-B.); (D.P.d.P.); (R.M.)
- Laboratory of Cellular Neurobiology, Department of Basic Medical Sciences, Faculty of Health Sciences, University of La Laguna, 38200 Tenerife, Spain
- Associate Research Unit ULL-CSIC “Membrane Physiology and Biophysics in Neurodegenerative and Cancer Diseases”, 38200 Tenerife, Spain
| |
Collapse
|
29
|
Aleksic M, Rajagopal R, de-Ávila R, Spriggs S, Gilmour N. The skin sensitization adverse outcome pathway: exploring the role of mechanistic understanding for higher tier risk assessment. Crit Rev Toxicol 2024; 54:69-91. [PMID: 38385441 DOI: 10.1080/10408444.2024.2308816] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Accepted: 12/19/2023] [Indexed: 02/23/2024]
Abstract
For over a decade, the skin sensitization Adverse Outcome Pathway (AOP) has served as a useful framework for development of novel in chemico and in vitro assays for use in skin sensitization hazard and risk assessment. Since its establishment, the AOP framework further fueled the existing efforts in new assay development and stimulated a plethora of activities with particular focus on validation, reproducibility and interpretation of individual assays and combination of assay outputs for use in hazard/risk assessment. In parallel, research efforts have also accelerated in pace, providing new molecular and dynamic insight into key events leading to sensitization. In light of novel hypotheses emerging from over a decade of focused research effort, mechanistic evidence relating to the key events in the skin sensitization AOP may complement the tools currently used in risk assessment. We reviewed the recent advances unraveling the complexity of molecular events in sensitization and signpost the most promising avenues for further exploration and development of useful assays.
Collapse
Affiliation(s)
- Maja Aleksic
- Safety and Environmental Assurance Centre, Unilever, Sharnbrook, UK
| | - Ramya Rajagopal
- Safety and Environmental Assurance Centre, Unilever, Sharnbrook, UK
| | - Renato de-Ávila
- Safety and Environmental Assurance Centre, Unilever, Sharnbrook, UK
| | - Sandrine Spriggs
- Safety and Environmental Assurance Centre, Unilever, Sharnbrook, UK
| | - Nicola Gilmour
- Safety and Environmental Assurance Centre, Unilever, Sharnbrook, UK
| |
Collapse
|
30
|
Shahtout JL, Eshima H, Ferrara PJ, Maschek JA, Cox JE, Drummond MJ, Funai K. Inhibition of the skeletal muscle Lands cycle ameliorates weakness induced by physical inactivity. J Cachexia Sarcopenia Muscle 2024; 15:319-330. [PMID: 38123161 PMCID: PMC10834354 DOI: 10.1002/jcsm.13406] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Revised: 10/12/2023] [Accepted: 11/15/2023] [Indexed: 12/23/2023] Open
Abstract
BACKGROUND Lipid hydroperoxides (LOOH) have been implicated in skeletal muscle atrophy with age and disuse. Lysophosphatidylcholine acyltransferase 3 (LPCAT3), an enzyme of the Lands cycle, conjugates a polyunsaturated fatty acyl chain to a lysophospholipid to form a polyunsaturated fatty acid containing phospholipid (PUFA-PL) molecule, providing substrates for LOOH propagation. Previous studies suggest that inhibition of the Lands cycle is an effective strategy to suppress LOOH. Mice with skeletal muscle-specific tamoxifen-inducible knockout of LPCAT3 (LPCAT3-MKO) were utilized to determine if muscle-specific attenuation of LOOH may alleviate muscle atrophy and weakness with disuse. METHODS LPCAT3-MKO and control mice underwent 7 days of sham or hindlimb unloading (HU model) to study muscle mass and force-generating capacity. LOOH was assessed by quantifying 4-hydroxynonenal (4-HNE)-conjugated peptides. Quantitative PCR and lipid mass spectrometry were used to validate LPCAT3 deletion. RESULTS Seven days of HU was sufficient to induce muscle atrophy and weakness concomitant to a ~2-fold increase in 4-HNE (P = 0.0069). Deletion of LPCAT3 reversed HU-induced increase in muscle 4-HNE (P = 0.0256). No difference was found in body mass, body composition, or caloric intake between genotypes. The soleus (SOL) and plantaris (PLANT) muscles of the LPCAT3-MKO mice experienced ~15% and ~40% less atrophy than controls, respectively. (P = 0.0011 and P = 0.0265). Type I and IIa SOL myofibers experienced a ~40% decrease in cross sectional area (CSA), which was attenuated to only 15% in the LPCAT3-MKO mice (P = 0.0170 and P = 0.0411, respectively). Strikingly, SOL muscles were fully protected and extensor digitorum longus (EDL) muscles experienced a ~35% protection from HU-induced reduction in force-generating capacity in the LPCAT3-MKO mice compared with controls (P < 0.0001 for both muscles). CONCLUSIONS Our findings demonstrate that attenuation of skeletal muscle lipid hydroperoxides is sufficient to restore its function, in particular a protection from reduction in muscle specific force. Our findings suggest muscle lipid peroxidation contributes to atrophy and weakness induced by disuse in mice.
Collapse
Affiliation(s)
- Justin L. Shahtout
- Diabetes and Metabolism Research CenterUniversity of UtahSalt Lake CityUtahUSA
- Department of Physical Therapy and Athletic TrainingUniversity of UtahSalt Lake CityUtahUSA
| | - Hiroaki Eshima
- Diabetes and Metabolism Research CenterUniversity of UtahSalt Lake CityUtahUSA
- Molecular Medicine ProgramUniversity of UtahSalt Lake CityUtahUSA
- Nagasaki International UniversitySaseboJapan
| | - Patrick J. Ferrara
- Diabetes and Metabolism Research CenterUniversity of UtahSalt Lake CityUtahUSA
- Molecular Medicine ProgramUniversity of UtahSalt Lake CityUtahUSA
| | - J. Alan Maschek
- Diabetes and Metabolism Research CenterUniversity of UtahSalt Lake CityUtahUSA
- Metabolomics, Mass Spectrometry, and Proteomics CoreUniversity of UtahSalt Lake CityUtahUSA
| | - James E. Cox
- Diabetes and Metabolism Research CenterUniversity of UtahSalt Lake CityUtahUSA
- Metabolomics, Mass Spectrometry, and Proteomics CoreUniversity of UtahSalt Lake CityUtahUSA
- Department of BiochemistryUniversity of UtahSalt Lake CityUtahUSA
| | - Micah J. Drummond
- Diabetes and Metabolism Research CenterUniversity of UtahSalt Lake CityUtahUSA
- Department of Physical Therapy and Athletic TrainingUniversity of UtahSalt Lake CityUtahUSA
- Molecular Medicine ProgramUniversity of UtahSalt Lake CityUtahUSA
- Department of Nutrition and Integrative PhysiologyUniversity of UtahSalt Lake CityUtahUSA
| | - Katsuhiko Funai
- Diabetes and Metabolism Research CenterUniversity of UtahSalt Lake CityUtahUSA
- Department of Physical Therapy and Athletic TrainingUniversity of UtahSalt Lake CityUtahUSA
- Molecular Medicine ProgramUniversity of UtahSalt Lake CityUtahUSA
- Department of BiochemistryUniversity of UtahSalt Lake CityUtahUSA
- Department of Nutrition and Integrative PhysiologyUniversity of UtahSalt Lake CityUtahUSA
| |
Collapse
|
31
|
Sørensen M, Pershagen G, Thacher JD, Lanki T, Wicki B, Röösli M, Vienneau D, Cantuaria ML, Schmidt JH, Aasvang GM, Al-Kindi S, Osborne MT, Wenzel P, Sastre J, Fleming I, Schulz R, Hahad O, Kuntic M, Zielonka J, Sies H, Grune T, Frenis K, Münzel T, Daiber A. Health position paper and redox perspectives - Disease burden by transportation noise. Redox Biol 2024; 69:102995. [PMID: 38142584 PMCID: PMC10788624 DOI: 10.1016/j.redox.2023.102995] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Revised: 12/07/2023] [Accepted: 12/10/2023] [Indexed: 12/26/2023] Open
Abstract
Transportation noise is a ubiquitous urban exposure. In 2018, the World Health Organization concluded that chronic exposure to road traffic noise is a risk factor for ischemic heart disease. In contrast, they concluded that the quality of evidence for a link to other diseases was very low to moderate. Since then, several studies on the impact of noise on various diseases have been published. Also, studies investigating the mechanistic pathways underlying noise-induced health effects are emerging. We review the current evidence regarding effects of noise on health and the related disease-mechanisms. Several high-quality cohort studies consistently found road traffic noise to be associated with a higher risk of ischemic heart disease, heart failure, diabetes, and all-cause mortality. Furthermore, recent studies have indicated that road traffic and railway noise may increase the risk of diseases not commonly investigated in an environmental noise context, including breast cancer, dementia, and tinnitus. The harmful effects of noise are related to activation of a physiological stress response and nighttime sleep disturbance. Oxidative stress and inflammation downstream of stress hormone signaling and dysregulated circadian rhythms are identified as major disease-relevant pathomechanistic drivers. We discuss the role of reactive oxygen species and present results from antioxidant interventions. Lastly, we provide an overview of oxidative stress markers and adverse redox processes reported for noise-exposed animals and humans. This position paper summarizes all available epidemiological, clinical, and preclinical evidence of transportation noise as an important environmental risk factor for public health and discusses its implications on the population level.
Collapse
Affiliation(s)
- Mette Sørensen
- Work, Environment and Cancer, Danish Cancer Institute, Copenhagen, Denmark; Department of Natural Science and Environment, Roskilde University, Denmark.
| | - Göran Pershagen
- Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Jesse Daniel Thacher
- Division of Occupational and Environmental Medicine, Department of Laboratory Medicine, Lund University, Lund, Sweden
| | - Timo Lanki
- Department of Health Security, Finnish Institute for Health and Welfare, Kuopio, Finland; School of Medicine, University of Eastern Finland, Kuopio, Finland; Department of Environmental and Biological Sciences, University of Eastern Finland, Kuopio, Finland
| | - Benedikt Wicki
- Department of Epidemiology and Public Health, Swiss Tropical and Public Health Institute, Allschwil, Switzerland; University of Basel, Basel, Switzerland
| | - Martin Röösli
- Department of Epidemiology and Public Health, Swiss Tropical and Public Health Institute, Allschwil, Switzerland; University of Basel, Basel, Switzerland
| | - Danielle Vienneau
- Department of Epidemiology and Public Health, Swiss Tropical and Public Health Institute, Allschwil, Switzerland; University of Basel, Basel, Switzerland
| | - Manuella Lech Cantuaria
- Work, Environment and Cancer, Danish Cancer Institute, Copenhagen, Denmark; Research Unit for ORL - Head & Neck Surgery and Audiology, Odense University Hospital & University of Southern Denmark, Odense, Denmark
| | - Jesper Hvass Schmidt
- Research Unit for ORL - Head & Neck Surgery and Audiology, Odense University Hospital & University of Southern Denmark, Odense, Denmark
| | - Gunn Marit Aasvang
- Department of Air Quality and Noise, Norwegian Institute of Public Health, Oslo, Norway
| | - Sadeer Al-Kindi
- Department of Medicine, University Hospitals, Harrington Heart & Vascular Institute, Case Western Reserve University, 11100 Euclid Ave, Cleveland, OH, 44106, USA
| | - Michael T Osborne
- Cardiovascular Imaging Research Center, Massachusetts General Hospital, Boston, MA, USA; Division of Cardiology, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Philip Wenzel
- Department of Cardiology, Cardiology I, University Medical Center Mainz, Mainz, Germany; German Center for Cardiovascular Research (DZHK), Partner Site Rhine-Main, Mainz, Germany; Center for Thrombosis and Hemostasis, University Medical Center Mainz, Mainz, Germany
| | - Juan Sastre
- Department of Physiology, Faculty of Pharmacy, University of Valencia, Spain
| | - Ingrid Fleming
- Institute for Vascular Signalling, Centre for Molecular Medicine, Goethe University, Frankfurt Am Main, Germany; German Center of Cardiovascular Research (DZHK), Partner Site RheinMain, Frankfurt, Germany
| | - Rainer Schulz
- Institute of Physiology, Faculty of Medicine, Justus-Liebig University, Gießen, 35392, Gießen, Germany
| | - Omar Hahad
- Department of Cardiology, Cardiology I, University Medical Center Mainz, Mainz, Germany; German Center for Cardiovascular Research (DZHK), Partner Site Rhine-Main, Mainz, Germany
| | - Marin Kuntic
- Department of Cardiology, Cardiology I, University Medical Center Mainz, Mainz, Germany; German Center for Cardiovascular Research (DZHK), Partner Site Rhine-Main, Mainz, Germany
| | - Jacek Zielonka
- Department of Biophysics, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Helmut Sies
- Institute for Biochemistry and Molecular Biology I, Faculty of Medicine, Heinrich Heine University Düsseldorf, Düsseldorf, Germany; Leibniz Research Institute for Environmental Medicine, Düsseldorf, Germany
| | - Tilman Grune
- Department of Molecular Toxicology, German Institute of Human Nutrition Potsdam-Rehbruecke, Nuthetal, Germany; DZHK (German Center for Cardiovascular Research), Partner Site Berlin, Berlin, Germany; German Center for Diabetes Research (DZD), München-Neuherberg, Germany
| | - Katie Frenis
- Hematology/Oncology, Boston Children's Hospital and Harvard Medical School, Boston, MA, USA; Stem Cell Program, Boston Children's Hospital, Boston, MA, USA
| | - Thomas Münzel
- Department of Cardiology, Cardiology I, University Medical Center Mainz, Mainz, Germany; German Center for Cardiovascular Research (DZHK), Partner Site Rhine-Main, Mainz, Germany; Center for Thrombosis and Hemostasis, University Medical Center Mainz, Mainz, Germany
| | - Andreas Daiber
- Department of Cardiology, Cardiology I, University Medical Center Mainz, Mainz, Germany; German Center for Cardiovascular Research (DZHK), Partner Site Rhine-Main, Mainz, Germany; Center for Thrombosis and Hemostasis, University Medical Center Mainz, Mainz, Germany.
| |
Collapse
|
32
|
Wang Y, Yang J, Zhang Y, Zhou J. Focus on Mitochondrial Respiratory Chain: Potential Therapeutic Target for Chronic Renal Failure. Int J Mol Sci 2024; 25:949. [PMID: 38256023 PMCID: PMC10815764 DOI: 10.3390/ijms25020949] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Revised: 12/26/2023] [Accepted: 01/09/2024] [Indexed: 01/24/2024] Open
Abstract
The function of the respiratory chain is closely associated with kidney function, and the dysfunction of the respiratory chain is a primary pathophysiological change in chronic kidney failure. The incidence of chronic kidney failure caused by defects in respiratory-chain-related genes has frequently been overlooked. Correcting abnormal metabolic reprogramming, rescuing the "toxic respiratory chain", and targeting the clearance of mitochondrial reactive oxygen species are potential therapies for treating chronic kidney failure. These treatments have shown promising results in slowing fibrosis and inflammation progression and improving kidney function in various animal models of chronic kidney failure and patients with chronic kidney disease (CKD). The mitochondrial respiratory chain is a key target worthy of attention in the treatment of chronic kidney failure. This review integrated research related to the mitochondrial respiratory chain and chronic kidney failure, primarily elucidating the pathological status of the mitochondrial respiratory chain in chronic kidney failure and potential therapeutic drugs. It provided new ideas for the treatment of kidney failure and promoted the development of drugs targeting the mitochondrial respiratory chain.
Collapse
Affiliation(s)
| | | | | | - Jianhua Zhou
- Department of Pediatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science & Technology, Wuhan 430030, China; (Y.W.); (J.Y.); (Y.Z.)
| |
Collapse
|
33
|
Favero G, Golic I, Arnaboldi F, Cappella A, Korac A, Monsalve M, Stacchiotti A, Rezzani R. Cardiometabolic Changes in Sirtuin1-Heterozygous Mice on High-Fat Diet and Melatonin Supplementation. Int J Mol Sci 2024; 25:860. [PMID: 38255934 PMCID: PMC10815439 DOI: 10.3390/ijms25020860] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Revised: 01/04/2024] [Accepted: 01/08/2024] [Indexed: 01/24/2024] Open
Abstract
A hypercaloric fatty diet predisposes an individual to metabolic syndrome and cardiovascular complications. Sirtuin1 (SIRT1) belongs to the class III histone deacetylase family and sustains anabolism, mitochondrial biogenesis, and fat distribution. Epididymal white adipose tissue (eWAT) is involved in inflammation, whilst interscapular brown adipose tissue (iBAT) drives metabolism in obese rodents. Melatonin, a pineal indoleamine, acting as a SIRT1 modulator, may alleviate cardiometabolic damage. In the present study, we morphologically characterized the heart, eWAT, and iBAT in male heterozygous SIRT1+/- mice (HET mice) on a high-fat diet (60%E lard) versus a standard rodent diet (8.5% E fat) and drinking melatonin (10 mg/kg) for 16 weeks. Wild-type (WT) male C57Bl6/J mice were similarly fed for comparison. Cardiomyocyte fibrosis and endoplasmic reticulum (ER) stress response worsened in HET mice on a high-fat diet vs. other groups. Lipid peroxidation, ER, and mitochondrial stress were assessed by 4 hydroxy-2-nonenal (4HNE), glucose-regulated protein78 (GRP78), CCAA/enhancer-binding protein homologous protein (CHOP), heat shock protein 60 (HSP60), and mitofusin2 immunostainings. Ultrastructural analysis indicated the prevalence of atypical inter-myofibrillar mitochondria with short, misaligned cristae in HET mice on a lard diet despite melatonin supplementation. Abnormal eWAT adipocytes, crown-like inflammatory structures, tumor necrosis factor alpha (TNFα), and iBAT whitening characterized HET mice on a hypercaloric fatty diet and were maintained after melatonin supply. All these data suggest that melatonin's mechanism of action is strictly linked to full SIRT1 expression, which is required for the exhibition of effective antioxidant and anti-inflammatory properties.
Collapse
Affiliation(s)
- Gaia Favero
- Anatomy and Physiopathology Division, Department of Clinical and Experimental Sciences, University of Brescia, Viale Europa 11, 25123 Brescia, Italy; (G.F.); (R.R.)
- Interdipartimental University Center of Research “Adaption and Regeneration of Tissues and Organs (ARTO)”, University of Brescia, 25123 Brescia, Italy
| | - Igor Golic
- Center for Electron Microscopy, Faculty of Biology, University of Belgrade, Studentski trg 16, 11000 Belgrade, Serbia; (I.G.); (A.K.)
| | - Francesca Arnaboldi
- Department of Biomedical Sciences for Health, University of Milan, Via Mangiagalli 31, 20133 Milan, Italy; (F.A.); (A.C.)
| | - Annalisa Cappella
- Department of Biomedical Sciences for Health, University of Milan, Via Mangiagalli 31, 20133 Milan, Italy; (F.A.); (A.C.)
- U.O. Laboratorio di Morfologia Umana Applicata, IRCCS Policlinico San Donato, San Donato Milanese, 20097 Milan, Italy
| | - Aleksandra Korac
- Center for Electron Microscopy, Faculty of Biology, University of Belgrade, Studentski trg 16, 11000 Belgrade, Serbia; (I.G.); (A.K.)
| | - Maria Monsalve
- Instituto de Investigaciones Biomedicas “Alberto Sols” (CSIC-UAM), 28029 Madrid, Spain;
| | - Alessandra Stacchiotti
- Department of Biomedical Sciences for Health, University of Milan, Via Mangiagalli 31, 20133 Milan, Italy; (F.A.); (A.C.)
- U.O. Laboratorio di Morfologia Umana Applicata, IRCCS Policlinico San Donato, San Donato Milanese, 20097 Milan, Italy
| | - Rita Rezzani
- Anatomy and Physiopathology Division, Department of Clinical and Experimental Sciences, University of Brescia, Viale Europa 11, 25123 Brescia, Italy; (G.F.); (R.R.)
- Interdipartimental University Center of Research “Adaption and Regeneration of Tissues and Organs (ARTO)”, University of Brescia, 25123 Brescia, Italy
- Italian Society for the Study of Orofacial Pain (Società Italiana Studio Dolore Orofacciale—SISDO), 25123 Brescia, Italy
| |
Collapse
|
34
|
Moldogazieva NT, Zavadskiy SP, Astakhov DV, Terentiev AA. Lipid peroxidation: Reactive carbonyl species, protein/DNA adducts, and signaling switches in oxidative stress and cancer. Biochem Biophys Res Commun 2023; 687:149167. [PMID: 37939506 DOI: 10.1016/j.bbrc.2023.149167] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2023] [Revised: 10/15/2023] [Accepted: 10/26/2023] [Indexed: 11/10/2023]
Abstract
Under the exposure of lipids to reactive oxygen species (ROS), lipid peroxidation proceeds non-enzymatically and generates an extremely heterogeneous mixture of reactive carbonyl species (RCS). Among them, HNE, HHE, MDA, methylglyoxal, glyoxal, and acrolein are the most studied and/or abundant ones. Over the last decades, significant progress has been achieved in understanding mechanisms of RCS generation, protein/DNA adduct formation, and their identification and quantification in biological samples. In our review, we critically discuss the advancements in understanding the roles of RCS-induced protein/DNA modifications in signaling switches to provide adaptive cell response under physiological and oxidative stress conditions. At non-toxic concentrations, RCS modify susceptible Cys residue in c-Src to activate MAPK signaling and Cys, Lys, and His residues in PTEN to cause its reversible inactivation, thereby stimulating PI3K/PKB(Akt) pathway. RCS toxic concentrations cause irreversible Cys modifications in Keap1 and IKKβ followed by stabilization of Nrf2 and activation of NF-κB, respectively, for their nuclear translocation and antioxidant gene expression. Dysregulation of these mechanisms causes diseases including cancer. Alterations in RCS, RCS detoxifying enzymes, RCS-modified protein/DNA adducts, and signaling pathways have been implicated in various cancer types.
Collapse
Affiliation(s)
- Nurbubu T Moldogazieva
- Department of Pharmacology, A.P. Nelyubin Institute of Pharmacy, I.M. Sechenov First Moscow State Medical University, 119991, 8 Trubetskaya Street, Moscow, Russia.
| | - Sergey P Zavadskiy
- Department of Pharmacology, A.P. Nelyubin Institute of Pharmacy, I.M. Sechenov First Moscow State Medical University, 119991, 8 Trubetskaya Street, Moscow, Russia
| | - Dmitry V Astakhov
- Department of Biochemistry, Institute of Biodesign and Complex Systems Modelling, I.M. Sechenov First Moscow State Medical University, 119991, 8 Trubetskaya Str., Moscow, Russia
| | - Alexander A Terentiev
- Department of Biochemistry and Molecular Biology, N.I. Pirogov Russian National Research Medical University, 117997, 1 Ostrovityanov Street, Moscow, Russia
| |
Collapse
|
35
|
Kang S, Noh Y, Oh SJ, Yoon HJ, Im S, Kwon HT, Pak YK. Neuroprotective Effects of Aldehyde-Reducing Composition in an LPS-Induced Neuroinflammation Model of Parkinson's Disease. Molecules 2023; 28:7988. [PMID: 38138478 PMCID: PMC10745824 DOI: 10.3390/molecules28247988] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Revised: 11/30/2023] [Accepted: 12/06/2023] [Indexed: 12/24/2023] Open
Abstract
Parkinson's disease (PD) is a complex neurodegenerative disease in which neuroinflammation and oxidative stress interact to contribute to pathogenesis. This study investigates the in vivo neuroprotective effects of a patented yeast extract lysate in a lipopolysaccharide (LPS)-induced neuroinflammation model. The yeast extract lysate, named aldehyde-reducing composition (ARC), exhibited potent antioxidant and anti-aldehyde activities in vitro. Oral administration of ARC at 10 or 20 units/kg/day for 3 days prior to intraperitoneal injection of LPS (10 mg/kg) effectively preserved dopaminergic neurons in the substantia nigra (SN) and striatum by preventing LPS-induced cell death. ARC also normalized the activation of microglia and astrocytes in the SN, providing further evidence for its neuroprotective properties. In the liver, ARC downregulated the LPS-induced increase in inflammatory cytokines and reversed the LPS-induced decrease in antioxidant-related genes. These findings indicate that ARC exerts potent antioxidant, anti-aldehyde, and anti-inflammatory effects in vivo, suggesting its potential as a disease-modifying agent for the prevention and treatment of neuroinflammation-related diseases, including Parkinson's disease.
Collapse
Affiliation(s)
- Sora Kang
- Department of Neuroscience, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea; (S.K.); (H.J.Y.)
- Picoentech Co., Ltd., Seongnam-si 13201, Gyeong gi-do, Republic of Korea; (Y.N.); (H.T.K.)
| | - Youngjin Noh
- Picoentech Co., Ltd., Seongnam-si 13201, Gyeong gi-do, Republic of Korea; (Y.N.); (H.T.K.)
| | - Seung Jun Oh
- Department of Biomedical Sciences, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea; (S.J.O.); (S.I.)
| | - Hye Ji Yoon
- Department of Neuroscience, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea; (S.K.); (H.J.Y.)
| | - Suyeol Im
- Department of Biomedical Sciences, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea; (S.J.O.); (S.I.)
| | - Hung Taeck Kwon
- Picoentech Co., Ltd., Seongnam-si 13201, Gyeong gi-do, Republic of Korea; (Y.N.); (H.T.K.)
| | - Youngmi Kim Pak
- Department of Neuroscience, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea; (S.K.); (H.J.Y.)
- Department of Biomedical Sciences, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea; (S.J.O.); (S.I.)
- Department of Physiology, College of Medicine, Kyung Hee University, Seoul 02447, Republic of Korea
| |
Collapse
|
36
|
Denolly S, Stukalov A, Barayeu U, Rosinski AN, Kritsiligkou P, Joecks S, Dick TP, Pichlmair A, Bartenschlager R. Zika virus remodelled ER membranes contain proviral factors involved in redox and methylation pathways. Nat Commun 2023; 14:8045. [PMID: 38052817 DOI: 10.1038/s41467-023-43665-6] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2023] [Accepted: 11/16/2023] [Indexed: 12/07/2023] Open
Abstract
Zika virus (ZIKV) has emerged as a global health issue, yet neither antiviral therapy nor a vaccine are available. ZIKV is an enveloped RNA virus, replicating in the cytoplasm in close association with ER membranes. Here, we isolate ER membranes from ZIKV-infected cells and determine their proteome. Forty-six host cell factors are enriched in ZIKV remodeled membranes, several of these having a role in redox and methylation pathways. Four proteins are characterized in detail: thioredoxin reductase 1 (TXNRD1) contributing to folding of disulfide bond containing proteins and modulating ZIKV secretion; aldo-keto reductase family 1 member C3 (AKR1C3), regulating capsid protein abundance and thus, ZIKV assembly; biliverdin reductase B (BLVRB) involved in ZIKV induced lipid peroxidation and increasing stability of viral transmembrane proteins; adenosylhomocysteinase (AHCY) indirectly promoting m6A methylation of ZIKV RNA by decreasing the level of S- adenosyl homocysteine and thus, immune evasion. These results highlight the involvement of redox and methylation enzymes in the ZIKV life cycle and their accumulation at virally remodeled ER membranes.
Collapse
Affiliation(s)
- Solène Denolly
- Heidelberg University, Medical Faculty Heidelberg, Department of Infectious Diseases, Molecular Virology, Center for Integrative Infectious Disease Research, 69120, Heidelberg, Germany
| | - Alexey Stukalov
- Technical University of Munich, School of Medicine, Institute of Virology, 81675, Munich, Germany
| | - Uladzimir Barayeu
- Division of Redox Regulation, German Cancer Research Center (DKFZ), DKFZ-ZMBH Alliance, Heidelberg, Germany
- Faculty of Biosciences, Heidelberg University, Heidelberg, Germany
| | - Alina N Rosinski
- Heidelberg University, Medical Faculty Heidelberg, Department of Infectious Diseases, Molecular Virology, Center for Integrative Infectious Disease Research, 69120, Heidelberg, Germany
| | - Paraskevi Kritsiligkou
- Division of Redox Regulation, German Cancer Research Center (DKFZ), DKFZ-ZMBH Alliance, Heidelberg, Germany
| | - Sebastian Joecks
- Heidelberg University, Medical Faculty Heidelberg, Department of Infectious Diseases, Molecular Virology, Center for Integrative Infectious Disease Research, 69120, Heidelberg, Germany
| | - Tobias P Dick
- Division of Redox Regulation, German Cancer Research Center (DKFZ), DKFZ-ZMBH Alliance, Heidelberg, Germany
- Faculty of Biosciences, Heidelberg University, Heidelberg, Germany
| | - Andreas Pichlmair
- Technical University of Munich, School of Medicine, Institute of Virology, 81675, Munich, Germany
- German Center for Infection Research (DZIF), Munich Partner Site, Munich, Germany
| | - Ralf Bartenschlager
- Heidelberg University, Medical Faculty Heidelberg, Department of Infectious Diseases, Molecular Virology, Center for Integrative Infectious Disease Research, 69120, Heidelberg, Germany.
- Division Virus-Associated Carcinogenesis, German Cancer Research Center (DKFZ), Heidelberg, Germany.
| |
Collapse
|
37
|
Senturk A, Alver A, Karkucak M, Küçük M, Ahmadi Rendi T. Oxidative modification of carbonic anhydrase by peroxynitrite trigger immune response in mice and rheumatic disease patients. Am J Med Sci 2023; 366:438-448. [PMID: 37678670 DOI: 10.1016/j.amjms.2023.09.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Revised: 08/11/2023] [Accepted: 09/04/2023] [Indexed: 09/09/2023]
Abstract
BACKGROUND Carbonic anhydrases (CA) are metalloenzymes with wide tissue distribution, involved in many important physiological processes, and in some rheumatic diseases, autoantibodies are formed against these enzymes. Recent studies have suggested that oxidative stress triggers anti-CA antibody formation. In this study, we aimed to investigate the effects of modification with oxidative/nitrosative stress end products on CA antigenicity in mice and the relationship between the modified CA autoantibodies and oxidant-antioxidant status in patients with rheumatoid arthritis (RA) and Sjögren's syndrome (SjS). METHODS CA I and CA II isoenzymes were isolated from human erythrocytes and modified with 4-hydroxynonenal (4-HNE), malondialdehyde (MDA), and peroxynitrite (PN). Balb-c mice were immunized with these agents to determine the effects of modification on CA antigenicity. The autoantibody titers of modified CA isoenzymes were detected in patients. In addition MDA, 4-HNE, 3-nitrotyrosine (3-NT), superoxide dismutase (SOD), and glutathione peroxidase (GSH-Px) activities were measured to assess the oxidant-antioxidant status in patients. RESULTS Modifications of carbonic anhydrase with oxidative stress end products, HNE, MDA and PN, lead to alterations in the immune response to these enzymes in mice. It was found that HNE and MDA decreased the antigenicity while PN increased. In addition, PN-modified CA autoantibody levels were found to be significantly different in both RA and SjS patients compared to their controls (p<0.05). CONCLUSIONS PN modifications can also trigger an immune response against CA isoenzymes in mice, and PN-modified CA I and CA II autoantibody titers were found at a significantly high level in both RA and SjS patients.
Collapse
Affiliation(s)
- Ayse Senturk
- Macka Vocational School, Karadeniz Technical University, Trabzon 61750, Türkiye.
| | - Ahmet Alver
- Faculty of Medicine, Department of Medical Biochemistry, Karadeniz Technical University, Trabzon, Türkiye
| | - Murat Karkucak
- Faculty of Medicine, Department of Physical Medicine and Rehabilitation, Karadeniz Technical University, Trabzon, Türkiye
| | - Murat Küçük
- Faculty of Science, Department of Chemistry, Karadeniz Technical University, Trabzon, Türkiye
| | - Taghi Ahmadi Rendi
- Graduate School of Medical Science, Department of Medical Biochemistry, Karadeniz Technical University, Trabzon, Türkiye; Faculty of Medicine, Department of Medical Biochemistry, Istanbul University, Istanbul, Turkey
| |
Collapse
|
38
|
Guo X, Dilidaxi D, Li L, Wang C, Ma X, Sang F, Pei G, Li W. Aspirin protects human trophoblast HTR-8/SVneo cells from H 2O 2-Induced oxidative stress via NADPH/ROS pathway. Placenta 2023; 144:55-63. [PMID: 37995441 DOI: 10.1016/j.placenta.2023.11.008] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/30/2023] [Revised: 11/12/2023] [Accepted: 11/15/2023] [Indexed: 11/25/2023]
Abstract
INTRODUCTION Pre-eclampsia (PE) is a pregnancy complication that can lead to maternal, fetal, and neonatal deaths in clinical practice. Accumulation of trophoblastic reactive oxygen species (ROS), which could result in oxidative stress and cell apoptosis, is considered to play an important role in PE pathology. It has been reported that aspirin has a positive effect on PE treatment in high-risk pregnant women. METHODS In vitro, extravillous trophoblast cell line (HTR-8/SVneo) were treated with hydrogen peroxide (H2O2, 150 μM) after the presence of aspirin (90 and 120 μM) with or without GKT137831 (a Nox4 inhibitor, 20 μM). A series of experiments including CCK-8 assays, flow cytometry, biochemical testing, and Western Blotting etc. verified the protective effects and potential mechanisms of aspirin against oxidative stress-induced damage in PE. RESULTS Our results demonstrated that H2O2 induces oxidative stress and apoptosis in HTR8/SVneo cells. However, aspirin pretreatment rescue cell viability and reduce LDH activity of HTR-8/SVneo cells. Aspirin can suppress the ROS overproduction and MDA level while increase SOD content and CAT activity. In addition, aspirin pretreatment significantly alleviated cell apoptosis and suppressed the expression of Nox4 and its subunits (p22phox and p47phox) at protein and mRNA levels. The above results were more obvious after the combination of aspirin with GKT137831. DISCUSSION This study demonstrated that aspirin protects human trophoblasts against H2O2-induced oxidative stress and cell apoptosis via suppressing NADPH/ROS pathway. These findings provide novel insights for the application of aspirin as a protective and curative agent against PE.
Collapse
Affiliation(s)
- Xin Guo
- Department of Pharmacy, Urumqi Youai Hospital, Urumqi, 830000, China
| | - Dinareer Dilidaxi
- Department of Pharmacy, Urumqi Youai Hospital, Urumqi, 830000, China
| | - Lihua Li
- Department of Pharmacy, Urumqi Youai Hospital, Urumqi, 830000, China
| | - Chunyan Wang
- Department of Pharmacy, Urumqi Youai Hospital, Urumqi, 830000, China
| | - Xiaoqing Ma
- Department of Pharmacy, Urumqi Youai Hospital, Urumqi, 830000, China
| | - Fei Sang
- Department of Pharmacy, Urumqi Youai Hospital, Urumqi, 830000, China
| | - Guizhen Pei
- Department of Pharmacy, Xinjiang Production & Construction Corps Hospital, Urumqi, 830000, China.
| | - Wei Li
- Department of Pharmacy, Urumqi Youai Hospital, Urumqi, 830000, China.
| |
Collapse
|
39
|
Gao Y, Zhang Y, Liu W, Zhang N, Gao Q, Shangguan J, Li N, Zhao Y, Jia Y. Danggui Buxue decoction alleviates cyclophosphamide-induced myelosuppression by regulating β-hydroxybutyric acid metabolism and suppressing oxidative stress. PHARMACEUTICAL BIOLOGY 2023; 61:710-721. [PMID: 37096658 PMCID: PMC10132245 DOI: 10.1080/13880209.2023.2201606] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 05/03/2023]
Abstract
CONTEXT Danggui Buxue Decoction (DBD) is an effective complementary medicine in alleviating myelosuppression after chemotherapy (MAC). However, its mechanism of action is elusive. OBJECTIVE To illustrate that regulating β-hydroxybutyric acid (β-OHB) metabolism and suppressing oxidative stress could be a potential mechanism of action for DBD in alleviating MAC. MATERIALS AND METHODS After HPLC quantification and dose testing (3, 6 and 10 g/kg, gavage) of DBD, Sprague-Dawley rats were divided into control, cyclophosphamide (CTX) (30 mg/kg CTX for 5 days, intraperitoneal administration) and CTX + DBD groups (6 g/kg DBD for 14 days, gavage). Blood cell counts, thigh bone histological examination, β-OHB levels, oxidative stress indices and HDAC1 activity were tested. The biological function of β-OHB was verified in vitro (hBMSC cells were incubated in culture mediums that contained 40 μM CTX and β-OHB in 0, 1, 2.5, 5, 10 mM) and in vivo (MAC rat model, 3 g/kg β-OHB for 14 days, gavage). RESULTS Rats in the CTX + DBD group showed upregulated blood cell counts (118-243%), β-OHB levels (495 nmol/mL in blood, 122 nmol/mg in marrow supernatant) and downregulated HDAC1 activity (59%), and oxidative stress indices (60-85%). In vitro, 5 mM β-OHB improved hBMSC cell migration (123%) and proliferation (131%). In vivo, rats treated with 3 g/kg β-OHB showed upregulated blood cell counts (121-182%) and downregulated HDAC1 activity (64%) and oxidative stress indices (65-83%). DISCUSSION AND CONCLUSIONS DBD, a traditional Chinese medicine, alleviates MAC by intervening in β-OHB metabolism and oxidative stress.
Collapse
Affiliation(s)
- Yiqiao Gao
- School of Pharmacy, Xinxiang Medical University, Xinxiang, P. R. China
- Xinxiang Key Laboratory of Clinical Psychopharmacology, Xinxiang Medical University, Xinxiang, P. R. China
- CONTACT Yiqiao Gao
| | - Yixin Zhang
- School of Pharmacy, Xinxiang Medical University, Xinxiang, P. R. China
- Xinxiang Key Laboratory of Clinical Psychopharmacology, Xinxiang Medical University, Xinxiang, P. R. China
| | - Wei Liu
- School of Pharmacy, Xinxiang Medical University, Xinxiang, P. R. China
| | - Nan Zhang
- School of Pharmacy, Xinxiang Medical University, Xinxiang, P. R. China
| | - Qinghe Gao
- School of Pharmacy, Xinxiang Medical University, Xinxiang, P. R. China
| | | | - Na Li
- School of Pharmacy, Xinxiang Medical University, Xinxiang, P. R. China
- Xinxiang Key Laboratory of Clinical Psychopharmacology, Xinxiang Medical University, Xinxiang, P. R. China
| | - Ying Zhao
- School of Pharmacy, Xinxiang Medical University, Xinxiang, P. R. China
- Xinxiang Key Laboratory of Clinical Psychopharmacology, Xinxiang Medical University, Xinxiang, P. R. China
| | - Yanlong Jia
- School of Pharmacy, Xinxiang Medical University, Xinxiang, P. R. China
- Yanlong Jia School of Pharmacy, Xinxiang Medical University, Xinxiang, Henan453003, P. R. China
| |
Collapse
|
40
|
Ivarsson J, Ferrara F, Vallese A, Guiotto A, Colella S, Pecorelli A, Valacchi G. Comparison of Pollutant Effects on Cutaneous Inflammasomes Activation. Int J Mol Sci 2023; 24:16674. [PMID: 38068996 PMCID: PMC10706824 DOI: 10.3390/ijms242316674] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2023] [Revised: 11/13/2023] [Accepted: 11/16/2023] [Indexed: 12/18/2023] Open
Abstract
The skin is the outermost layer of the body and, therefore, is exposed to a variety of stressors, such as environmental pollutants, known to cause oxinflammatory reactions involved in the exacerbation of several skin conditions. Today, inflammasomes are recognized as important modulators of the cutaneous inflammatory status in response to air pollutants and ultraviolet (UV) light exposure. In this study, human skin explants were exposed to the best-recognized air pollutants, such as microplastics (MP), cigarette smoke (CS), diesel engine exhaust (DEE), ozone (O3), and UV, for 1 or 4 days, to explore how each pollutant can differently modulate markers of cutaneous oxinflammation. Exposure to environmental pollutants caused an altered oxidative stress response, accompanied by increased DNA damage and signs of premature skin aging. The effect of specific pollutants being able to exert different inflammasomes pathways (NLRP1, NLRP3, NLRP6, and NLRC4) was also investigated in terms of scaffold formation and cell pyroptosis. Among all environmental pollutants, O3, MP, and UV represented the main pollutants affecting cutaneous redox homeostasis; of note, the NLRP1 and NLRP6 inflammasomes were the main ones modulated by these outdoor stressors, suggesting their role as possible molecular targets in preventing skin disorders and the inflammaging events associated with environmental pollutant exposure.
Collapse
Affiliation(s)
- John Ivarsson
- Department of Food, Bioprocessing and Nutrition Sciences, Plants for Human Health Institute, NC Research Campus, NC State University, Kannapolis, NC 28081, USA;
| | - Francesca Ferrara
- Department of Chemical, Pharmaceuticals and Agricultural Sciences, University of Ferrara, 44121 Ferrara, Italy;
| | - Andrea Vallese
- Department of Animal Sciences, Plants for Human Health Institute, NC Research Campus, NC State University, Kannapolis, NC 28081, USA; (A.V.); (A.G.); (A.P.)
- Department of Environmental Sciences and Prevention, University of Ferrara, 44121 Ferrara, Italy
| | - Anna Guiotto
- Department of Animal Sciences, Plants for Human Health Institute, NC Research Campus, NC State University, Kannapolis, NC 28081, USA; (A.V.); (A.G.); (A.P.)
- Department of Neuroscience and Rehabilitation, University of Ferrara, 44121 Ferrara, Italy
| | - Sante Colella
- Department of Biotechnology, Chemistry and Pharmaceutical Sciences, University of Siena, 53100 Siena, Italy;
| | - Alessandra Pecorelli
- Department of Animal Sciences, Plants for Human Health Institute, NC Research Campus, NC State University, Kannapolis, NC 28081, USA; (A.V.); (A.G.); (A.P.)
- Department of Environmental Sciences and Prevention, University of Ferrara, 44121 Ferrara, Italy
| | - Giuseppe Valacchi
- Department of Animal Sciences, Plants for Human Health Institute, NC Research Campus, NC State University, Kannapolis, NC 28081, USA; (A.V.); (A.G.); (A.P.)
- Department of Environmental Sciences and Prevention, University of Ferrara, 44121 Ferrara, Italy
- Department of Food and Nutrition, Kyung Hee University, Seoul 26723, Republic of Korea
| |
Collapse
|
41
|
Cortez NE, Lanzi CR, Vahmani P, Matsukuma K, Mackenzie GG. Hepatic safety profile of pancreatic cancer‑bearing mice fed a ketogenic diet in combination with gemcitabine. Oncol Lett 2023; 26:479. [PMID: 37818128 PMCID: PMC10561147 DOI: 10.3892/ol.2023.14067] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Accepted: 09/06/2023] [Indexed: 10/12/2023] Open
Abstract
Ketogenic diets (KDs) are actively being evaluated for their potential anticancer effects. Although KDs are generally considered safe, their safety profile when combined with chemotherapy remains unknown. It is known that a KD enhances the anticancer effect of gemcitabine (2',2'-difluoro-2'-deoxycytidine) in LSL-KrasLSL-G12D/+Trp53R172H/+Pdx-1-Cre (KPC) tumor-bearing mice. In the present study, whether a KD in combination with gemcitabine affected the liver safety profile in KPC mice was evaluated. For this purpose, male and female pancreatic tumor-bearing KPC mice were allocated to a control diet (CD; % kcal: 20% fat, 65% carbohydrate, 15% protein) + gemcitabine [control plus gemcitabine group (CG)] or a KD (% kcal: 84% fat, 15% protein, 1% carbohydrate) + gemcitabine [ketogenic plus gemcitabine group (KG)] for two months. After two months of treatment, no significant differences in body weight were observed between CGs and KGs. Moreover, the KD did not significantly alter the serum protein expression levels of liver enzymes, including aspartate aminotransferase, alanine aminotransferase and alkaline phosphatase. In addition, the KD did not alter markers of liver-lipid accumulation as well as serum cholesterol and triglyceride levels, compared with the CG-treated group. Upon histologic examination, steatosis was rare, with no notable differences between treatment groups. When examining liver fatty acid composition, KD treatment significantly increased the content of saturated fatty acids and significantly decreased levels of cis-monounsaturated fatty acids compared with the CG. Finally, the KD did not affect liver markers of inflammation and oxidative stress, nor the protein expression levels of enzymes involved in ketone bodies, such as 3-hydroxy-3-methylglutaryl-CoA lyase and hidroximetilglutaril-CoA sintasa, and glucose metabolism, such as hexokinase 2, pyruvate dehydrogenase and phosphofructokinase. In summary, a KD in combination with gemcitabine appears to be safe, with no apparent hepatotoxicity and these data support the further evaluation of a KD as an adjuvant dietary treatment for pancreatic cancer.
Collapse
Affiliation(s)
- Natalia E. Cortez
- Department of Nutrition, University of California, Davis, CA 95616, USA
| | | | - Payam Vahmani
- Department of Animal Science, University of California, Davis, CA 95616, USA
| | - Karen Matsukuma
- Department of Pathology and Laboratory Medicine, Davis Medical Center, University of California, Sacramento, CA 95817, USA
- University of California Davis Comprehensive Cancer Center, University of California, Sacramento, CA 95817, USA
| | - Gerardo G. Mackenzie
- Department of Nutrition, University of California, Davis, CA 95616, USA
- University of California Davis Comprehensive Cancer Center, University of California, Sacramento, CA 95817, USA
| |
Collapse
|
42
|
Cho YE, Kim Y, Kim SJ, Lee H, Hwang S. Overexpression of Interleukin-8 Promotes the Progression of Fatty Liver to Nonalcoholic Steatohepatitis in Mice. Int J Mol Sci 2023; 24:15489. [PMID: 37895168 PMCID: PMC10607753 DOI: 10.3390/ijms242015489] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2023] [Revised: 10/18/2023] [Accepted: 10/21/2023] [Indexed: 10/29/2023] Open
Abstract
Nonalcoholic steatohepatitis (NASH) is an advanced stage of fatty liver disease characterized by liver damage, inflammation, and fibrosis. Although neutrophil infiltration is consistently observed in the livers of patients with NASH, the precise role of neutrophil-recruiting chemokines and infiltrating neutrophils in NASH pathogenesis remains poorly understood. Here, we aimed to elucidate the role of neutrophil infiltration in the transition from fatty liver to NASH by examining hepatic overexpression of interleukin-8 (IL8), a major chemokine responsible for neutrophil recruitment in humans. Mice fed a high-fat diet (HFD) for 3 months developed fatty liver without concurrent liver damage, inflammation, and fibrosis. Subsequent infection with an adenovirus overexpressing human IL8 for an additional 2 weeks increased IL8 levels, neutrophil infiltration, and liver injury in mice. Mechanistically, IL8-induced liver injury was associated with the upregulation of components of the NADPH oxidase 2 complex, which participate in neutrophil oxidative burst. IL8-driven neutrophil infiltration promoted macrophage aggregate formation and upregulated the expression of chemokines and inflammatory cytokines. Notably, IL8 overexpression amplified factors associated with fibrosis, including collagen deposition and hepatic stellate cell activation, in HFD-fed mice. Collectively, hepatic overexpression of human IL8 promotes neutrophil infiltration and fatty liver progression to NASH in HFD-fed mice.
Collapse
Affiliation(s)
- Ye Eun Cho
- College of Pharmacy and Research Institute for Drug Development, Pusan National University, Busan 46241, Republic of Korea; (Y.E.C.); (Y.K.); (H.L.)
| | - Yeonsoo Kim
- College of Pharmacy and Research Institute for Drug Development, Pusan National University, Busan 46241, Republic of Korea; (Y.E.C.); (Y.K.); (H.L.)
| | - Seung-Jin Kim
- Department of Biochemistry, College of Natural Sciences, Kangwon National University, Chuncheon 24341, Republic of Korea;
| | - Haeseung Lee
- College of Pharmacy and Research Institute for Drug Development, Pusan National University, Busan 46241, Republic of Korea; (Y.E.C.); (Y.K.); (H.L.)
| | - Seonghwan Hwang
- College of Pharmacy and Research Institute for Drug Development, Pusan National University, Busan 46241, Republic of Korea; (Y.E.C.); (Y.K.); (H.L.)
- Laboratory of Liver Diseases, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD 20892, USA
| |
Collapse
|
43
|
Ji D, Luo M, Guo Y, Li Q, Kong L, Ge H, Wang Q, Song Q, Zeng X, Ma J, Wang Y, Meurer J, Chi W. Efficient scavenging of reactive carbonyl species in chloroplasts is required for light acclimation and fitness of plants. THE NEW PHYTOLOGIST 2023; 240:676-693. [PMID: 37545368 DOI: 10.1111/nph.19156] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Accepted: 07/03/2023] [Indexed: 08/08/2023]
Abstract
Reactive carbonyl species (RCS) derived from lipid peroxides can act as critical damage or signaling mediators downstream of reactive oxygen species by modifying target proteins. However, their biological effects and underlying mechanisms remain largely unknown in plants. Here, we have uncovered the mechanism by which the RCS 4-hydroxy-(E)-2-nonenal (HNE) participates in photosystem II (PSII) repair cycle of chloroplasts, a crucial process for maintaining PSII activity under high and changing light conditions. High Light Sensitive 1 (HLT1) is a potential NADPH-dependent reductase in chloroplasts. Deficiency of HLT1 had no impact on the growth of Arabidopsis plants under normal light conditions but increased sensitivity to high light, which resulted from a defective PSII repair cycle. In hlt1 plants, the accumulation of HNE-modified D1 subunit of PSII was observed, which did not affect D1 degradation but hampered the dimerization of repaired PSII monomers and reassembly of PSII supercomplexes on grana stacks. HLT1 is conserved in all photosynthetic organisms and has functions in overall growth and plant fitness in both Arabidopsis and rice under naturally challenging field conditions. Our work provides the mechanistic basis underlying RCS scavenging in light acclimation and suggests a potential strategy to improve plant productivity by manipulating RCS signaling in chloroplasts.
Collapse
Affiliation(s)
- Daili Ji
- Photosynthesis Research Center, Key Laboratory of Photobiology, Institute of Botany, Chinese Academy of Sciences, Beijing, 100093, China
| | - Manfei Luo
- Photosynthesis Research Center, Key Laboratory of Photobiology, Institute of Botany, Chinese Academy of Sciences, Beijing, 100093, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Yinjie Guo
- Photosynthesis Research Center, Key Laboratory of Photobiology, Institute of Botany, Chinese Academy of Sciences, Beijing, 100093, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Qiuxin Li
- Photosynthesis Research Center, Key Laboratory of Photobiology, Institute of Botany, Chinese Academy of Sciences, Beijing, 100093, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Lingxi Kong
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Haitao Ge
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Qi Wang
- Institute of Crop Cultivation and Tillage, Heilongjiang Academy of Agricultural Sciences, Harbin, 150086, China
| | - Qiulai Song
- Institute of Crop Cultivation and Tillage, Heilongjiang Academy of Agricultural Sciences, Harbin, 150086, China
| | - Xiannan Zeng
- Institute of Crop Cultivation and Tillage, Heilongjiang Academy of Agricultural Sciences, Harbin, 150086, China
| | - Jinfang Ma
- Photosynthesis Research Center, Key Laboratory of Photobiology, Institute of Botany, Chinese Academy of Sciences, Beijing, 100093, China
| | - Yingchun Wang
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Jörg Meurer
- Plant Molecular Biology, Faculty of Biology, Ludwig-Maximilians-University, D-82152, Planegg-Martinsried, Munich, Germany
| | - Wei Chi
- Photosynthesis Research Center, Key Laboratory of Photobiology, Institute of Botany, Chinese Academy of Sciences, Beijing, 100093, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
- The Innovative Academy of Seed Design, Chinese Academy of Sciences, Beijing, 100101, China
| |
Collapse
|
44
|
Zhang X, Hou L, Guo Z, Wang G, Xu J, Zheng Z, Sun K, Guo F. Lipid peroxidation in osteoarthritis: focusing on 4-hydroxynonenal, malondialdehyde, and ferroptosis. Cell Death Discov 2023; 9:320. [PMID: 37644030 PMCID: PMC10465515 DOI: 10.1038/s41420-023-01613-9] [Citation(s) in RCA: 51] [Impact Index Per Article: 25.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Revised: 08/11/2023] [Accepted: 08/16/2023] [Indexed: 08/31/2023] Open
Abstract
Osteoarthritis (OA) is a multifactorial and increasingly prevalent degenerative disease that affects the whole joint. The pathogenesis of OA is poorly understood and there is a lack of therapeutic interventions to reverse the pathological process of this disease. Accumulating studies have shown that the overproduction of reactive oxygen species (ROS) and ROS-induced lipid peroxidation are involved in the pathogenesis of OA. 4-Hydroxy-2-nonenal (4-HNE) and malondialdehyde (MDA) have received considerable attention for their role in cartilage degeneration and subchondral bone remodeling during OA development. Ferroptosis is a form of cell death characterized by a lack of control of membrane lipid peroxidation and recent studies have suggested that chondrocyte ferroptosis contributes to OA progression. In this review, we aim to discuss lipid peroxidation-derived 4-HNE and MDA in the progression of OA. In addition, the therapeutic potential for OA by controlling the accumulation of lipid peroxidation and inhibiting chondrocyte ferroptosis are discussed.
Collapse
Affiliation(s)
- Xiong Zhang
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China
| | - Liangcai Hou
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China
| | - Zhou Guo
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China
| | - Genchun Wang
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China
| | - Jingting Xu
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China
| | - Zehang Zheng
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China
| | - Kai Sun
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China.
| | - Fengjing Guo
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China.
| |
Collapse
|
45
|
Shahtout JL, Eshima H, Ferrara PJ, Maschek JA, Cox JE, Drummond MJ, Funai K. Inhibition of skeletal muscle Lands cycle ameliorates weakness induced by physical inactivity. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.07.25.550576. [PMID: 37546754 PMCID: PMC10402104 DOI: 10.1101/2023.07.25.550576] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/08/2023]
Abstract
Background Lipid hydroperoxides (LOOH) have been implicated in skeletal muscle atrophy with age and disuse. Lysophosphatidylcholine acyltransferase 3 (LPCAT3), an enzyme of Lands cycle, conjugates a polyunsaturated fatty acyl chain to a lysophospholipid (PUFA-PL) molecule, providing substrates for LOOH propagation. Previous studies suggest that inhibition of Lands cycle is an effective strategy to suppress LOOH. Mice with skeletal muscle-specific tamoxifen-inducible knockout of LPCAT3 (LPCAT3-MKO) were utilized to determine if muscle-specific attenuation of LOOH may alleviate muscle atrophy and weakness with disuse. Methods LPCAT3-MKO and control mice underwent 7 days of sham or hindlimb unloading (HU model) to study muscle mass and force-generating capacity. LOOH was assessed by quantifying 4-hydroxynonenal (4-HNE)-conjugated peptides. Quantitative PCR and lipid mass spectrometry were used to validate LPCAT3 deletion. Results 7 days of HU was sufficient to induce muscle atrophy and weakness concomitant to an increase in 4-HNE. Deletion of LPCAT3 reversed HU-induced increase in muscle 4HNE. No difference was found in body mass, body composition, or caloric intake between genotypes. The soleus (SOL) and plantaris (PLANT) muscles of the LPCAT3-MKO mice were partially protected from atrophy compared to controls, concomitant to attenuated decrease in cross-sectional areas in type I and IIa fibers. Strikingly, SOL and extensor digitorum longus (EDL) were robustly protected from HU-induced reduction in force-generating capacity in the LPCAT3-MKO mice compared to controls. Conclusion Our findings demonstrate that attenuation of muscle LOOH is sufficient to restore skeletal muscle function, in particular a protection from reduction in muscle specific force. Thus, muscle LOOH contributes to atrophy and weakness induced by HU in mice.
Collapse
Affiliation(s)
- Justin L. Shahtout
- Diabetes and Metabolism Research Center, University of Utah, Salt Lake City, UT, USA
- Department of Physical Therapy and Athletic Training, University of Utah, Salt Lake City, UT, USA
| | - Hiroaki Eshima
- Diabetes and Metabolism Research Center, University of Utah, Salt Lake City, UT, USA
- Molecular Medicine Program, University of Utah, Salt Lake City, UT, USA
- Nagasaki International University, Sasebo, Nagasaki, Japan
| | - Patrick J. Ferrara
- Diabetes and Metabolism Research Center, University of Utah, Salt Lake City, UT, USA
- Molecular Medicine Program, University of Utah, Salt Lake City, UT, USA
| | - J. Alan Maschek
- Diabetes and Metabolism Research Center, University of Utah, Salt Lake City, UT, USA
- Metabolomics, Mass Spectrometry, and Proteomics Core, University of Utah, Salt Lake City, UT. USA
| | - James E. Cox
- Diabetes and Metabolism Research Center, University of Utah, Salt Lake City, UT, USA
- Metabolomics, Mass Spectrometry, and Proteomics Core, University of Utah, Salt Lake City, UT. USA
- Department of Biochemistry, University of Utah, Salt Lake City, UT, USA
| | - Micah J. Drummond
- Diabetes and Metabolism Research Center, University of Utah, Salt Lake City, UT, USA
- Department of Physical Therapy and Athletic Training, University of Utah, Salt Lake City, UT, USA
- Molecular Medicine Program, University of Utah, Salt Lake City, UT, USA
- Department of Nutrition and Integrative Physiology, University of Utah, Salt Lake City, UT, USA
| | - Katsuhiko Funai
- Diabetes and Metabolism Research Center, University of Utah, Salt Lake City, UT, USA
- Department of Physical Therapy and Athletic Training, University of Utah, Salt Lake City, UT, USA
- Molecular Medicine Program, University of Utah, Salt Lake City, UT, USA
- Department of Nutrition and Integrative Physiology, University of Utah, Salt Lake City, UT, USA
| |
Collapse
|
46
|
Karan BM, Little K, Augustine J, Stitt AW, Curtis TM. Aldehyde Dehydrogenase and Aldo-Keto Reductase Enzymes: Basic Concepts and Emerging Roles in Diabetic Retinopathy. Antioxidants (Basel) 2023; 12:1466. [PMID: 37508004 PMCID: PMC10376360 DOI: 10.3390/antiox12071466] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Revised: 07/18/2023] [Accepted: 07/19/2023] [Indexed: 07/30/2023] Open
Abstract
Diabetic retinopathy (DR) is a complication of diabetes mellitus that can lead to vision loss and blindness. It is driven by various biochemical processes and molecular mechanisms, including lipid peroxidation and disrupted aldehyde metabolism, which contributes to retinal tissue damage and the progression of the disease. The elimination and processing of aldehydes in the retina rely on the crucial role played by aldehyde dehydrogenase (ALDH) and aldo-keto reductase (AKR) enzymes. This review article investigates the impact of oxidative stress, lipid-derived aldehydes, and advanced lipoxidation end products (ALEs) on the advancement of DR. It also provides an overview of the ALDH and AKR enzymes expressed in the retina, emphasizing their growing importance in DR. Understanding the relationship between aldehyde metabolism and DR could guide innovative therapeutic strategies to protect the retina and preserve vision in diabetic patients. This review, therefore, also explores various approaches, such as gene therapy and pharmacological compounds that have the potential to augment the expression and activity of ALDH and AKR enzymes, underscoring their potential as effective treatment options for DR.
Collapse
Affiliation(s)
- Burak Mugdat Karan
- Wellcome-Wolfson Institute for Experimental Medicine, Queen's University Belfast, Belfast BT7 1NN, UK
| | - Karis Little
- Wellcome-Wolfson Institute for Experimental Medicine, Queen's University Belfast, Belfast BT7 1NN, UK
| | - Josy Augustine
- Wellcome-Wolfson Institute for Experimental Medicine, Queen's University Belfast, Belfast BT7 1NN, UK
| | - Alan W Stitt
- Wellcome-Wolfson Institute for Experimental Medicine, Queen's University Belfast, Belfast BT7 1NN, UK
| | - Tim M Curtis
- Wellcome-Wolfson Institute for Experimental Medicine, Queen's University Belfast, Belfast BT7 1NN, UK
| |
Collapse
|
47
|
Nousis L, Kanavaros P, Barbouti A. Oxidative Stress-Induced Cellular Senescence: Is Labile Iron the Connecting Link? Antioxidants (Basel) 2023; 12:1250. [PMID: 37371980 DOI: 10.3390/antiox12061250] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2023] [Revised: 06/02/2023] [Accepted: 06/06/2023] [Indexed: 06/29/2023] Open
Abstract
Cellular senescence, a cell state characterized by a generally irreversible cell cycle arrest, is implicated in various physiological processes and a wide range of age-related pathologies. Oxidative stress, a condition caused by an imbalance between the production and the elimination of reactive oxygen species (ROS) in cells and tissues, is a common driver of cellular senescence. ROS encompass free radicals and other molecules formed as byproducts of oxygen metabolism, which exhibit varying chemical reactivity. A prerequisite for the generation of strong oxidizing ROS that can damage macromolecules and impair cellular function is the availability of labile (redox-active) iron, which catalyzes the formation of highly reactive free radicals. Targeting labile iron has been proven an effective strategy to counteract the adverse effects of ROS, but evidence concerning cellular senescence is sparse. In the present review article, we discuss aspects of oxidative stress-induced cellular senescence, with special attention to the potential implication of labile iron.
Collapse
Affiliation(s)
- Lambros Nousis
- Department of Hygiene and Epidemiology, Faculty of Medicine, School of Health Sciences, University of Ioannina, 45110 Ioannina, Greece
| | - Panagiotis Kanavaros
- Department of Anatomy-Histology-Embryology, Faculty of Medicine, School of Health Sciences, University of Ioannina, 45110 Ioannina, Greece
| | - Alexandra Barbouti
- Department of Anatomy-Histology-Embryology, Faculty of Medicine, School of Health Sciences, University of Ioannina, 45110 Ioannina, Greece
| |
Collapse
|
48
|
Abeer MI, Abdulhasan A, Haguar Z, Narayanaswami V. Isoform-specific modification of apolipoprotein E by 4-hydroxynonenal: protective role of apolipoprotein E3 against oxidative species. FEBS J 2023; 290:3006-3025. [PMID: 36661393 PMCID: PMC11296219 DOI: 10.1111/febs.16729] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Revised: 12/02/2022] [Accepted: 01/18/2023] [Indexed: 01/21/2023]
Abstract
High levels of 4-hydroxynonenal (HNE), arising from lipid peroxidation, and HNE-modified proteins have been identified in postmortem brains of ageing and Alzheimer's disease (AD) patients. The goal of this study is to understand the effect of HNE modification on the structure and function of recombinant apolipoprotein E3 (apoE3) and apolipoprotein E4 (apoE4), which play a critical role in brain cholesterol homeostasis. The two isoforms differ in a single amino acid at position 112: Cys in apoE3 and Arg in apoE4. Immunoblot with HNE-specific antibody indicates HNE modification of apoE3 and apoE4 with a major band at ~ 36 kDa, while LC-MS/MS revealed Michael addition at His140 (60-70% abundance) and His299 (3-5% abundance) in apoE3 and apoE4, and Cys112 adduct in apoE3 (75% abundance). Circular dichroism spectroscopy revealed no major differences in the overall secondary structure or helical content between unmodified and HNE-modified apoE. HNE modification did not affect their ability to promote cholesterol efflux from J774.1 macrophages. However, it led to a 3-fold decrease in their ability to bind lipids and 25-50% decrease in the ability of cerebral cortex endothelial cells to uptake lipoproteins bearing HNE-modified HNE-apoE3 or HNE-apoE4 as noted by fluorescence microscopy and flow cytometry. Taken together, the data indicate that HNE modification impairs lipid binding and cellular uptake of both isoforms, and that apoE3, bearing a Cys, offers a protective role by sequestering lipid peroxidation products that would otherwise cause indiscriminate damage to biomolecules. ApoE4, lacking Cys, is unable to protect against oxidative damage that is commensurate with ageing.
Collapse
Affiliation(s)
- Muhammad I Abeer
- Department of Chemistry and Biochemistry, California State University Long Beach, CA, USA
| | - Abbas Abdulhasan
- Department of Chemistry and Biochemistry, California State University Long Beach, CA, USA
| | - Zahraa Haguar
- Department of Chemistry and Biochemistry, California State University Long Beach, CA, USA
| | - Vasanthy Narayanaswami
- Department of Chemistry and Biochemistry, California State University Long Beach, CA, USA
| |
Collapse
|
49
|
Zhai C, Lonergan SM, Huff-Lonergan EJ, Johnson LG, Brown K, Prenni JE, N Nair M. Lipid Peroxidation Products Influence Calpain-1 Functionality In Vitro by Covalent Binding. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2023; 71:7836-7846. [PMID: 37167568 DOI: 10.1021/acs.jafc.3c01225] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/13/2023]
Abstract
The objective of the current study was to evaluate the effects of lipid peroxidation products, malondialdehyde (MDA), hexenal, and 4-hydroxynonenal (HNE), on calpain-1 function, and liquid chromatography and tandem mass spectrometry (LC-MS/MS) identification of adducts on calpain-1. Calpain-1 activity slightly increased after incubation with 100 μM MDA but not with 500 and 1000 μM MDA. However, calpain-1 activity was lowered by hexenal and HNE at 100, 500, and 1000 μM. No difference in calpain-1 autolysis was observed between the control and 1000 μM MDA. However, 1000 μM hexenal and HNE treatments slowed the calpain-1 autolysis. Adducts of MDA were detected on glutamine, arginine, lysine, histidine, and asparagine residues via Schiff base formation, while HNE adducts were detected on histidine, lysine, glutamine, and asparagine residues via Michael addition. These results are the first to demonstrate that lipid peroxidation products can impact calpain-1 activity in a concentration-dependent manner and may impact the development of meat tenderness postmortem.
Collapse
Affiliation(s)
- Chaoyu Zhai
- Department of Animal Science, University of Connecticut, Storrs, Connecticut 06269, United States
- Department of Animal Sciences, Colorado State University, Fort Collins, Colorado 80523, United States
| | - Steven M Lonergan
- Department of Animal Science, Iowa State University, Ames, Iowa 50011, United States
| | | | - Logan G Johnson
- Department of Animal Science, Iowa State University, Ames, Iowa 50011, United States
| | - Kitty Brown
- Analytical Resources Core-Bioanalysis & Omics, Colorado State University, Fort Collins, Colorado 80523, United States
| | - Jessica E Prenni
- Department of Horticulture and Landscape Architecture, Colorado State University, Fort Collins, Colorado 80523, United States
| | - Mahesh N Nair
- Department of Animal Sciences, Colorado State University, Fort Collins, Colorado 80523, United States
| |
Collapse
|
50
|
Bahou WF, Marchenko N, Nesbitt NM. Metabolic Functions of Biliverdin IXβ Reductase in Redox-Regulated Hematopoietic Cell Fate. Antioxidants (Basel) 2023; 12:antiox12051058. [PMID: 37237924 DOI: 10.3390/antiox12051058] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Revised: 04/19/2023] [Accepted: 04/27/2023] [Indexed: 05/28/2023] Open
Abstract
Cytoprotective heme oxygenases derivatize heme to generate carbon monoxide, ferrous iron, and isomeric biliverdins, followed by rapid NAD(P)H-dependent biliverdin reduction to the antioxidant bilirubin. Recent studies have implicated biliverdin IXβ reductase (BLVRB) in a redox-regulated mechanism of hematopoietic lineage fate restricted to megakaryocyte and erythroid development, a function distinct and non-overlapping from the BLVRA (biliverdin IXα reductase) homologue. In this review, we focus on recent progress in BLVRB biochemistry and genetics, highlighting human, murine, and cell-based studies that position BLVRB-regulated redox function (or ROS accumulation) as a developmentally tuned trigger that governs megakaryocyte/erythroid lineage fate arising from hematopoietic stem cells. BLVRB crystallographic and thermodynamic studies have elucidated critical determinants of substrate utilization, redox coupling and cytoprotection, and have established that inhibitors and substrates bind within the single-Rossmann fold. These advances provide unique opportunities for the development of BLVRB-selective redox inhibitors as novel cellular targets that retain potential for therapeutic applicability in hematopoietic (and other) disorders.
Collapse
Affiliation(s)
- Wadie F Bahou
- Department of Medicine, School of Medicine, Stony Brook University, Stony Brook, NY 11794, USA
| | - Natalia Marchenko
- Department of Pathology, Stony Brook University, Stony Brook, NY 11794, USA
| | - Natasha M Nesbitt
- Blood Cell Technologies, 25 Health Sciences Drive, Stony Brook, NY 11790, USA
| |
Collapse
|