1
|
Rojas-Solé C, Pinilla-González V, Lillo-Moya J, González-Fernández T, Saso L, Rodrigo R. Integrated approach to reducing polypharmacy in older people: exploring the role of oxidative stress and antioxidant potential therapy. Redox Rep 2024; 29:2289740. [PMID: 38108325 PMCID: PMC10732214 DOI: 10.1080/13510002.2023.2289740] [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: 12/19/2023] Open
Abstract
Increased life expectancy, attributed to improved access to healthcare and drug development, has led to an increase in multimorbidity, a key contributor to polypharmacy. Polypharmacy is characterised by its association with a variety of adverse events in the older persons. The mechanisms involved in the development of age-related chronic diseases are largely unknown; however, altered redox homeostasis due to ageing is one of the main theories. In this context, the present review explores the development and interaction between different age-related diseases, mainly linked by oxidative stress. In addition, drug interactions in the treatment of various diseases are described, emphasising that the holistic management of older people and their pathologies should prevail over the individual treatment of each condition.
Collapse
Affiliation(s)
- Catalina Rojas-Solé
- Molecular and Clinical Pharmacology Program, Institute of Biomedical Sciences, Faculty of Medicine, University of Chile, Santiago, Chile
| | - Víctor Pinilla-González
- Molecular and Clinical Pharmacology Program, Institute of Biomedical Sciences, Faculty of Medicine, University of Chile, Santiago, Chile
| | - José Lillo-Moya
- Molecular and Clinical Pharmacology Program, Institute of Biomedical Sciences, Faculty of Medicine, University of Chile, Santiago, Chile
| | - Tommy González-Fernández
- Molecular and Clinical Pharmacology Program, Institute of Biomedical Sciences, Faculty of Medicine, University of Chile, Santiago, Chile
| | - Luciano Saso
- Department of Physiology and Pharmacology “Vittorio Erspamer”, Faculty of Pharmacy and Medicine, Sapienza University, Rome, Italy
| | - Ramón Rodrigo
- Molecular and Clinical Pharmacology Program, Institute of Biomedical Sciences, Faculty of Medicine, University of Chile, Santiago, Chile
| |
Collapse
|
2
|
Farooqui AA, Farooqui T. Phospholipids, Sphingolipids, and Cholesterol-Derived Lipid Mediators and Their Role in Neurological Disorders. Int J Mol Sci 2024; 25:10672. [PMID: 39409002 PMCID: PMC11476704 DOI: 10.3390/ijms251910672] [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: 08/31/2024] [Revised: 10/01/2024] [Accepted: 10/01/2024] [Indexed: 10/20/2024] Open
Abstract
Neural membranes are composed of phospholipids, sphingolipids, cholesterol, and proteins. In response to cell stimulation or injury, the metabolism of lipids generates various lipid mediators, which perform many cellular functions. Thus, phospholipids release arachidonic acid or docosahexaenoic acid from the sn-2 position of the glycerol moiety by the action of phospholipases A2. Arachidonic acid is a precursor for prostaglandins, leukotrienes, thromboxane, and lipoxins. Among these mediators, prostaglandins, leukotrienes, and thromboxane produce neuroinflammation. In contrast, lipoxins produce anti-inflammatory and pro-resolving effects. Prostaglandins, leukotrienes, and thromboxane are also involved in cell proliferation, differentiation, blood clotting, and blood vessel permeability. In contrast, DHA-derived lipid mediators are called specialized pro-resolving lipid metabolites (SPMs). They include resolvins, protectins, and maresins. These mediators regulate immune function by producing anti-inflammatory, pro-resolving, and cell protective effects. Sphingolipid-derived metabolites are ceramide, ceramide1-phosphate, sphingosine, and sphingosine 1 phosphate. They regulate many cellular processes, including enzyme activities, cell migration and adhesion, inflammation, and immunity. Cholesterol is metabolized into hydroxycholesterols and 7-ketocholesterol, which not only disrupts membrane fluidity, but also promotes inflammation, oxidative stress, and apoptosis. These processes lead to cellular damage.
Collapse
Affiliation(s)
| | - Tahira Farooqui
- Department of Molecular and Cellular Biochemistry, The Ohio State University, Columbus, OH 43210, USA;
| |
Collapse
|
3
|
Whitehead B, Corbin D, Meadows E, Zhang N, Hollander JM, Karelina K, Weil ZM. Cerebral hypoperfusion exacerbates vascular dysfunction after traumatic brain injury. Exp Neurol 2024; 380:114907. [PMID: 39103029 DOI: 10.1016/j.expneurol.2024.114907] [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: 02/28/2024] [Revised: 06/17/2024] [Accepted: 07/29/2024] [Indexed: 08/07/2024]
Abstract
Traumatic brain injuries are extremely common, and although most patients recover from their injuries many TBI patients suffer prolonged symptoms and remain at a higher risk for developing cardiovascular disease and neurodegeneration. Moreover, it remains challenging to identify predictors of poor long-term outcomes. Here, we tested the hypothesis that preexisting cerebrovascular impairment exacerbates metabolic and vascular dysfunction and leads to worse outcomes after TBI. Male mice underwent a mild surgical reduction in cerebral blood flow using a model of bilateral carotid artery stenosis (BCAS) wherein steel microcoils were implanted around the carotid arteries. Then, 30 days post coil implantation, mice underwent TBI or sham surgery. Gene expression profiles, cerebral blood flow, metabolic function, oxidative damage, vascular health and angiogenesis were assessed. Single nuclei RNA sequencing of endothelial cells isolated from mice after TBI showed differential gene expression profiles after TBI and BCAS, that were further altered when mice underwent both challenges. TBI but not BCAS increased mitochondrial oxidative metabolism. Both BCAS and TBI decreased cerebrovascular responses to repeated whisker stimulation. BCAS induced oxidative damage and inflammation in the vasculature as well as loss of vascular density, and reduced the numbers of angiogenic tip cells. Finally, intravascular protein accumulation was increased among mice that experienced both BCAS and TBI. Overall, our findings reveal that a prior vascular impairment significantly alters the profile of vascular health and function of the cerebrovasculature, and when combined with TBI may result in worsened outcomes.
Collapse
Affiliation(s)
- Bailey Whitehead
- Department of Neuroscience and Rockefeller Neuroscience Institute, West Virginia University, Morgantown, WV, USA.
| | - Deborah Corbin
- Department of Neuroscience and Rockefeller Neuroscience Institute, West Virginia University, Morgantown, WV, USA
| | - Ethan Meadows
- Department of Neuroscience and Rockefeller Neuroscience Institute, West Virginia University, Morgantown, WV, USA; Department of Human Performance - Exercise Physiology, West Virginia University, Morgantown, WV, USA
| | - Ning Zhang
- Department of Neuroscience and Rockefeller Neuroscience Institute, West Virginia University, Morgantown, WV, USA
| | - John M Hollander
- Department of Neuroscience and Rockefeller Neuroscience Institute, West Virginia University, Morgantown, WV, USA; Department of Human Performance - Exercise Physiology, West Virginia University, Morgantown, WV, USA
| | - Kate Karelina
- Department of Neuroscience and Rockefeller Neuroscience Institute, West Virginia University, Morgantown, WV, USA
| | - Zachary M Weil
- Department of Neuroscience and Rockefeller Neuroscience Institute, West Virginia University, Morgantown, WV, USA
| |
Collapse
|
4
|
Lee S, Ohn J, Kang BM, Hwang ST, Kwon O. Activation of mitochondrial aldehyde dehydrogenase 2 promotes hair growth in human hair follicles. J Adv Res 2024; 64:237-247. [PMID: 37972887 PMCID: PMC11464481 DOI: 10.1016/j.jare.2023.11.014] [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: 04/09/2023] [Revised: 11/06/2023] [Accepted: 11/12/2023] [Indexed: 11/19/2023] Open
Abstract
INTRODUCTION Hair loss is a common phenomenon associated with various environmental and genetic factors. Mitochondrial dysfunction-induced oxidative stress has been recognized as a crucial determinant of hair follicle (HF) biology. Aldehyde dehydrogenase 2 (ALDH2) mitigates oxidative stress by detoxifying acetaldehyde. This study investigated the potential role of ALDH2 modulation in HF function and hair growth promotion. OBJECTIVES To evaluate the effects of ALDH2 activation on oxidative stress in HFs and hair growth promotion. METHODS The modulatory role of ALDH2 on HFs was investigated using an ALDH2 activator. ALDH2 expression in human HFs was evaluated through in vitro immunofluorescence staining. Ex vivo HF organ culture was employed to assess hair shaft elongation, while the fluorescence probe 2',7'- dichlorodihydrofluorescein diacetate was utilized to detect reactive oxygen species (ROS). An in vivo mouse model was used to determine whether ALDH2 activation induces anagen. RESULTS During the anagen phase, ALDH2 showed significantly higher intensity than that in the telogen phase, and its expression was primarily localized along the outer layer of HFs. ALDH2 activation promoted anagen phase induction by reducing ROS levels and enhancing reactive aldehyde clearance, which indicated that ALDH2 functions as a ROS scavenger within HFs. Moreover, ALDH2 activation upregulated Akt/GSK 3β/β-catenin signaling in HFs. CONCLUSIONS Our findings highlight the hair growth promotion effects of ALDH2 activation in HFs and its potential as a promising therapeutic approach for promoting anagen induction.
Collapse
Affiliation(s)
- Seunghee Lee
- Department of Dermatology, Seoul National University College of Medicine, Seoul 03080, South Korea; Laboratory of Cutaneous Aging and Hair Research, Biomedical Research Institute, Seoul National University Hospital, Seoul 03080, South Korea; Institute of Human-Environment Interface Biology, Medical Research Center, Seoul National University, Seoul 03080, South Korea; Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul 03080, South Korea
| | - Jungyoon Ohn
- Department of Dermatology, Seoul National University College of Medicine, Seoul 03080, South Korea; Laboratory of Cutaneous Aging and Hair Research, Biomedical Research Institute, Seoul National University Hospital, Seoul 03080, South Korea; Institute of Human-Environment Interface Biology, Medical Research Center, Seoul National University, Seoul 03080, South Korea
| | - Bo Mi Kang
- Department of Dermatology, Seoul National University College of Medicine, Seoul 03080, South Korea; Laboratory of Cutaneous Aging and Hair Research, Biomedical Research Institute, Seoul National University Hospital, Seoul 03080, South Korea; Institute of Human-Environment Interface Biology, Medical Research Center, Seoul National University, Seoul 03080, South Korea
| | | | - Ohsang Kwon
- Department of Dermatology, Seoul National University College of Medicine, Seoul 03080, South Korea; Laboratory of Cutaneous Aging and Hair Research, Biomedical Research Institute, Seoul National University Hospital, Seoul 03080, South Korea; Institute of Human-Environment Interface Biology, Medical Research Center, Seoul National University, Seoul 03080, South Korea; Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul 03080, South Korea.
| |
Collapse
|
5
|
Salama RM, Darwish SF, Yehia R, Sallam AA, Elmongy NF, Abd-Elgalil MM, El Wakeel SA. Lactoferrin alleviates gentamicin-induced acute kidney injury in rats by suppressing ferroptosis: Highlight on ACSL4, SLC7A11, NCOA4, FSP1 pathways and miR-378a-3p, LINC00618 expression. Food Chem Toxicol 2024; 193:115027. [PMID: 39357596 DOI: 10.1016/j.fct.2024.115027] [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: 06/04/2024] [Revised: 07/23/2024] [Accepted: 09/29/2024] [Indexed: 10/04/2024]
Abstract
The use of gentamicin (GNT) is associated with acute kidney injury (AKI). Ferroptosis is a newly recognized iron-dependent, non-apoptotic cell death that can lead to AKI. Lactoferrin (LF), an iron-binding glycoprotein, was previously reported to be renoprotective. Nonetheless, LF's impact on GNT-induced AKI and ferroptosis has not yet been investigated. Accordingly, we assessed the dose-dependent effect of LF on GNT-induced AKI and its influence on ferroptosis. Thirty-six male rats were allocated as control, LF, GNT (100 mg/kg/day, i.p.), and groups given LF (100, 200, and 300 mg/kg, p.o.) for 14 days prior concurrently with GNT (Day 8-14). The high dose of LF (300 mg/kg) showed better histopathological picture, higher creatinine clearance, reduced serum and urine levels of kidney injury markers when compared to the GNT group and the lower two doses. These nephroprotective effects of LF can be attributed to the observed reduction in renal ferrous iron, 4-HNE, and MDA, miR-378a-3p and ALOX15 expression, TFR1, NCOA4, and ACSL4 protein expression and the increased LINC00618 expression, GSH levels, GPX4, SLC7A11, and FSP1 protein expression. In conclusion, LF high dose was the most renoprotective against GNT-induced AKI, in which suppression of ferroptosis pathways was a likely contributor to its protective mechanism.
Collapse
Affiliation(s)
- Rania M Salama
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Misr International University (MIU), Cairo, Egypt.
| | - Samar F Darwish
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Badr University in Cairo (BUC), Cairo, Egypt.
| | - Rana Yehia
- Clinical Pharmacy, Faculty of Pharmacy, British University in Egypt (BUE), Cairo, Egypt.
| | - Al Aliaa Sallam
- Biochemistry Department, Faculty of Pharmacy, Badr University in Cairo (BUC), Cairo, Egypt; Biochemistry Department, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt.
| | - Noura F Elmongy
- Physiology Department, Damietta Faculty of Medicine, Al-Azhar University, Damietta, Egypt.
| | - Mona M Abd-Elgalil
- Histology and Cell Biology Department, Faculty of Medicine for Girls, Al-Azhar University, Cairo, Egypt.
| | - Sara A El Wakeel
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Misr International University (MIU), Cairo, Egypt.
| |
Collapse
|
6
|
Bogahawaththa S, Hara M, Furukawa T, Iwasaka C, Sawada T, Yamada G, Tokiya M, Kitagawa K, Miyake Y, Kido MA, Hirota Y, Matsumoto A. Asian Flush Gene Variant Enhances Cellular Immunogenicity of COVID-19 Vaccine: Prospective Observation in the Japanese General Population. Vaccines (Basel) 2024; 12:1015. [PMID: 39340045 PMCID: PMC11435883 DOI: 10.3390/vaccines12091015] [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: 07/18/2024] [Revised: 09/02/2024] [Accepted: 09/03/2024] [Indexed: 09/30/2024] Open
Abstract
We previously reported a reduced humoral immune response to the COVID-19 vaccines. Subsequently, we observed a lower susceptibility to COVID-19 in individuals carrying the ALDH2 rs671 variant through a web-based retrospective survey. Based on these findings, we hypothesized that rs671 variant was beneficial for cellular immunity against COVID-19. Using the IFN-γ enzyme-linked immunospot (ELISPOT) assay, we assessed cellular immunity before and after COVID-19 vaccination in two subcohorts of a previously reported cohort. Subcohort 1 (26 participants) had six repeated observations at baseline after one to three doses, whereas subcohort 2 (19 participants) had two observations before and after the third dose. ELISPOT counts at six months after the second dose increased from baseline in carriers of the rs671 variant but not in non-carriers. A positive effect of rs671 on ELISPOT counts was estimated using a mixed model (183 observations from 45 participants), including the random effect of subcohort, repeated measures, and fixed effects of vaccine type, age, sex, height, lifestyle, steroid use, and allergic disease. There was no association between ELISPOT counts and specific IgG levels, suggesting a limitation in estimating protective potential by humoral response. Our sequential observational studies suggest a beneficial effect of the rs671 variant in SARS-CoV-2 infection via enhanced cellular immune response, providing a potential basis for optimizing preventive measures and drug development.
Collapse
Affiliation(s)
- Sudarma Bogahawaththa
- Laboratory of Biochemistry, Department of Applied Biochemistry and Food Science, Faculty of Agriculture, Saga University, Honjo, Saga 840-8502, Japan;
- Department of Social and Environmental Medicine, Saga University, Nabeshima, Saga 840-8501, Japan;
| | - Megumi Hara
- Department of Preventive Medicine, Faculty of Medicine, Saga University, Nabeshima, Saga 840-8501, Japan; (M.H.); (T.F.); (C.I.)
| | - Takuma Furukawa
- Department of Preventive Medicine, Faculty of Medicine, Saga University, Nabeshima, Saga 840-8501, Japan; (M.H.); (T.F.); (C.I.)
- Clinical Research Center, Saga University Hospital, Nabeshima, Saga 840-8501, Japan
| | - Chiharu Iwasaka
- Department of Preventive Medicine, Faculty of Medicine, Saga University, Nabeshima, Saga 840-8501, Japan; (M.H.); (T.F.); (C.I.)
- Department of Physical Activity Research, National Institutes of Biomedical Innovation, Health and Nutrition, KENTO Innovation Park NK Building, 3–17, Senriokashinmachi, Settsu 566-0002, Japan
| | - Takeshi Sawada
- Department of Histology and Neuroanatomy, Faculty of Medicine, Saga University, Nabeshima, Saga 840-8501, Japan; (T.S.); (M.A.K.)
| | - Goki Yamada
- United Graduate School of Agriculture, Kagoshima University, Kagoshima 890-0065, Japan;
| | - Mikiko Tokiya
- Department of Social and Environmental Medicine, Saga University, Nabeshima, Saga 840-8501, Japan;
| | - Kyoko Kitagawa
- Division of Ultrastructural Cell Biology, Department of Anatomy, Faculty of Medicine, University of Miyazaki, Miyazaki 889-1692, Japan;
| | - Yasunobu Miyake
- Division of Molecular and Cellular Immunoscience, Department of Biomolecular Sciences, Faculty of Medicine, Saga University, 5-1-1 Nabeshima, Saga 849-8501, Japan;
| | - Mizuho Aoki Kido
- Department of Histology and Neuroanatomy, Faculty of Medicine, Saga University, Nabeshima, Saga 840-8501, Japan; (T.S.); (M.A.K.)
| | - Yoshio Hirota
- Clinical Epidemiology Research Center, SOUSEIKAI Medical Group (Medical Co., LTA), Fukuoka 813-0017, Japan;
| | - Akiko Matsumoto
- Department of Social and Environmental Medicine, Saga University, Nabeshima, Saga 840-8501, Japan;
| |
Collapse
|
7
|
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
|
8
|
Ko J, Jang S, Jang S, Park S, Yi J, Choi DK, Kim S, Kim MO, Lim SG, Ryoo ZY. Glucose-dependent insulinotropic polypeptide (GIP) alleviates ferroptosis in aging-induced brain damage through the Epac/Rap1 signaling pathway. BMB Rep 2024; 57:417-423. [PMID: 39219045 PMCID: PMC11444989] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Revised: 05/30/2024] [Accepted: 07/24/2024] [Indexed: 09/04/2024] Open
Abstract
Glucose-dependent insulinotropic polypeptide (GIP), a 42-aminoacid hormone, exerts multifaceted effects in physiology, most notably in metabolism, obesity, and inflammation. Its significance extends to neuroprotection, promoting neuronal proliferation, maintaining physiological homeostasis, and inhibiting cell death, all of which play a crucial role in the context of neurodegenerative diseases. Through intricate signaling pathways involving its cognate receptor (GIPR), a member of the G protein-coupled receptors, GIP maintains cellular homeostasis and regulates a defense system against ferroptosis, an essential process in aging. Our study, utilizing GIP-overexpressing mice and in vitro cell model, elucidates the pivotal role of GIP in preserving neuronal integrity and combating age-related damage, primarily through the Epac/Rap1 pathway. These findings shed light on the potential of GIP as a therapeutic target for the pathogenesis of ferroptosis in neurodegenerative diseases and aging. [BMB Reports 2024; 57(9): 417-423].
Collapse
Affiliation(s)
- Jiwon Ko
- Institute of Life Science and Biotechnology, Kyungpook National University, Daegu 41566, Korea
| | - Soyoung Jang
- School of Life Science and Biotechnology, College of Natural Sciences, BK21 FOUR KNU Creative BioResearch Group, Kyungpook National University, Daegu 41566, Korea
| | - Soyeon Jang
- Institute of Life Science and Biotechnology, Kyungpook National University, Daegu 41566, Korea
| | - Song Park
- Division of Animal Science, Gyeongsang National University, Jinju 52828, Korea
- Institute of Agriculture and Life Science (IALS), Gyeongsang National University, Jinju 52828, Korea
| | - Junkoo Yi
- School of Animal Life Convergence Science, Hankyong National University, Anseong 17579, Korea
| | - Dong Kyu Choi
- School of Life Science and Biotechnology, College of Natural Sciences, BK21 FOUR KNU Creative BioResearch Group, Kyungpook National University, Daegu 41566, Korea
| | - Seonggon Kim
- Preclinical Research Center, Daegu-Gyeongbuk Medical Innovation Foundation, Daegu 41061, Korea
| | - Myoung Ok Kim
- Department of Animal Science and Biotechnology, Kyungpook National University, Sangju 37224, Korea
| | - Su-Geun Lim
- Institute of Life Science and Biotechnology, Kyungpook National University, Daegu 41566, Korea
| | - Zae Young Ryoo
- School of Life Science and Biotechnology, College of Natural Sciences, BK21 FOUR KNU Creative BioResearch Group, Kyungpook National University, Daegu 41566, Korea
| |
Collapse
|
9
|
Luiz Antonio E, de Oliveira HA, Albuquerque-Pontes GM, Teixeira ILA, Yoshizaki AP, Dos Santos LFN, Leal-Junior ECP, Tucci PJF, Serra AJ. Examining the impact of varying low-level laser dose on cardiac failure. Photochem Photobiol 2024. [PMID: 39126163 DOI: 10.1111/php.14012] [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: 05/16/2024] [Revised: 07/26/2024] [Accepted: 07/27/2024] [Indexed: 08/12/2024]
Abstract
Low-level laser therapy (LLLT) has been targeted as a promising tool that can mitigate post-infarction cardiac remodeling. However, there is no gold standard energy delivered to the heart and few studies have evaluated the impact of LLLT on cardiac performance. This study evaluated effects of repeated LLLT applications with different energies delivered to the infarcted myocardium. Echocardiography and hemodynamic measurements were applied to evaluate left ventricular (LV) performance in rats with large infarcts. ELISA, Western blot and biochemical assays were used to assess LV inflammation and oxidative stress. An 830-nm Laser Photon III semiconductor aluminum gallium arsenide diode (DMC, São Carlos, SP, Brazil) was applied transthoracically three times a week for 4 weeks based on the energy (i.e., 10J, 20J, and 40J; respectively). LLLT on 10J and 20J had a similar action in attenuating pulmonary congestion and myocardial fibrosis. Moreover, 10J and 20J attenuated LV end-diastolic pressure and improved +dP/dt and -dP/dt. All LLLT groups had lower levels of inflammatory mediators, but only the 10J group had normalized oxidative stress. All LLLT doses improved superoxide dismutase levels; however, only the 20J group showed a high content of the catalase. There was a lower level of sarcoplasmic/endoplasmic reticulum Ca2+ ATPase 2a in the infarcted myocardium, which it was normalized in the 20J and 40J groups. A higher phospholamban content was found in the 10J group. This study supports the beneficial LLLT role post-infarction. Apparently, the 10J and 20J doses show to be chosen for clinical translation.
Collapse
Affiliation(s)
- Ednei Luiz Antonio
- Laboratory of Physiology and Cardiac Pathophysiology, Department of Medicine, Federal University of São Paulo, São Paulo, Brazil
| | - Helenita Antonia de Oliveira
- Laboratory of Physiology and Cardiac Pathophysiology, Department of Medicine, Federal University of São Paulo, São Paulo, Brazil
| | - Gianna Móes Albuquerque-Pontes
- Laboratory of Physiology and Cardiac Pathophysiology, Department of Medicine, Federal University of São Paulo, São Paulo, Brazil
| | - Ighor Luiz Azevedo Teixeira
- Laboratory of Physiology and Cardiac Pathophysiology, Department of Medicine, Federal University of São Paulo, São Paulo, Brazil
| | - Amanda Pereira Yoshizaki
- Laboratory of Physiology and Cardiac Pathophysiology, Department of Medicine, Federal University of São Paulo, São Paulo, Brazil
| | | | | | - Paulo José Ferreira Tucci
- Laboratory of Physiology and Cardiac Pathophysiology, Department of Medicine, Federal University of São Paulo, São Paulo, Brazil
| | - Andrey Jorge Serra
- Laboratory of Physiology and Cardiac Pathophysiology, Department of Medicine, Federal University of São Paulo, São Paulo, Brazil
| |
Collapse
|
10
|
Feng L, Sun J, Xia L, Shi Q, Hou Y, Zhang L, Li M, Fan C, Sun B. Ferroptosis mechanism and Alzheimer's disease. Neural Regen Res 2024; 19:1741-1750. [PMID: 38103240 PMCID: PMC10960301 DOI: 10.4103/1673-5374.389362] [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: 05/08/2023] [Revised: 07/05/2023] [Accepted: 09/09/2023] [Indexed: 12/18/2023] Open
Abstract
Regulated cell death is a genetically determined form of programmed cell death that commonly occurs during the development of living organisms. This process plays a crucial role in modulating homeostasis and is evolutionarily conserved across a diverse range of living organisms. Ferroptosis is a classic regulatory mode of cell death. Extensive studies of regulatory cell death in Alzheimer's disease have yielded increasing evidence that ferroptosis is closely related to the occurrence, development, and prognosis of Alzheimer's disease. This review summarizes the molecular mechanisms of ferroptosis and recent research advances in the role of ferroptosis in Alzheimer's disease. Our findings are expected to serve as a theoretical and experimental foundation for clinical research and targeted therapy for Alzheimer's disease.
Collapse
Affiliation(s)
- Lina Feng
- Shandong Key Laboratory of TCM Multi-Target Intervention and Disease Control, the Second Affiliated Hospital of Shandong First Medical University, Taian, Shandong Province, China
| | - Jingyi Sun
- Shandong Key Laboratory of TCM Multi-Target Intervention and Disease Control, the Second Affiliated Hospital of Shandong First Medical University, Taian, Shandong Province, China
| | - Ling Xia
- Shandong Key Laboratory of TCM Multi-Target Intervention and Disease Control, the Second Affiliated Hospital of Shandong First Medical University, Taian, Shandong Province, China
| | - Qiang Shi
- Shandong Key Laboratory of TCM Multi-Target Intervention and Disease Control, the Second Affiliated Hospital of Shandong First Medical University, Taian, Shandong Province, China
| | - Yajun Hou
- Shandong Key Laboratory of TCM Multi-Target Intervention and Disease Control, the Second Affiliated Hospital of Shandong First Medical University, Taian, Shandong Province, China
| | - Lili Zhang
- Department of Internal Medicine, Taian Traffic Hospital, Taian, Shandong Province, China
| | - Mingquan Li
- Department of Neurology, the Third Affiliated Clinical Hospital of Changchun University of Chinese Medicine, Changchun, Jilin Province, China
| | - Cundong Fan
- Shandong Key Laboratory of TCM Multi-Target Intervention and Disease Control, the Second Affiliated Hospital of Shandong First Medical University, Taian, Shandong Province, China
| | - Baoliang Sun
- Shandong Key Laboratory of TCM Multi-Target Intervention and Disease Control, the Second Affiliated Hospital of Shandong First Medical University, Taian, Shandong Province, China
| |
Collapse
|
11
|
Ivarsson J, Bennett A, Ferrara F, Strauch R, Vallase A, Iorizzo M, Pecorelli A, Lila MA, Valacchi G. Gut-derived wild blueberry phenolic acid metabolites modulate extrinsic cutaneous damage. Food Funct 2024; 15:7849-7864. [PMID: 38962816 DOI: 10.1039/d4fo01874e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/05/2024]
Abstract
As the first line of defense, the skin is equipped with various physiological mechanisms positioned to prevent incoming oxidative damage from numerous environmental insults. With persistent exposure to the environment, understanding ways to augment the skin defenses is paramount in protecting from premature aging. In this study, we investigated the ability of five dietary phenolic metabolites, typically found in the bloodstream after wild blueberry consumption, to successfully defend the skin from UV light exposure in a novel ex vivo co-culture model of human skin explants and primary endothelial cells. Skin explants, placed in transwell inserts, were exposed to UV, and subsequently co-cultured with endothelial cells. When the endothelial cells had been pretreated with the bioactive metabolites at physiological concentrations (hippuric acid 3000 nM, isoferulic acid 1000 nM, salicylic acid 130 nM, benzoic acid 900 nM, α-hydroxyhippuric acid 400 nM) cutaneous damage was prevented on the co-cultured with UV-challenged skin explants. Co-culture with non-pretreated endothelial cells did not protect skin explants. Specifically, the pretreatment was able to reduce skin lipid peroxidation (measured as 4-hydroxynonenal protein adducts), and pro-inflammatory enzymes such as cyclooxygenase 2 (COX-2) and NADPH oxidase 4 (NOX-4). Furthermore, pretreatment with the metabolites prevented UV-induced release of inflammatory cytokines such as IL-1β and IL-8 as well as nitric oxides (NO) levels. In addition, the metabolites showed an impressive ability to prevent the loss of cutaneous structural proteins including involucrin and collagen type 1. Of note, endothelial cells cultured with UV exposed skin explants exhibited increased oxidative stress demonstrated by heme oxygenase-1 (HO-1) up-regulation which was significantly prevented in the metabolite treated models. These findings highlight the ability of dietary polyphenolic metabolites to improve cutaneous defenses against extrinsic stressors.
Collapse
Affiliation(s)
- John Ivarsson
- Department of Animal Science, Plants for Human Health Institute, NC Research Campus, NC State University, Kannapolis, NC 28081, USA.
- Department of Food Bioprocessing & Nutrition Sciences, Plants for Human Health Institute, NC Research Campus, NC State University, Kannapolis, NC 28081, USA
| | - Abby Bennett
- Department of Food Bioprocessing & Nutrition Sciences, Plants for Human Health Institute, NC Research Campus, NC State University, Kannapolis, NC 28081, USA
| | - Francesca Ferrara
- Department of Chemical, Pharmaceutical and Agricultural Sciences, University of Ferrara, 44121 Ferrara, Italy
| | - Renee Strauch
- Department of Food Bioprocessing & Nutrition Sciences, Plants for Human Health Institute, NC Research Campus, NC State University, Kannapolis, NC 28081, USA
| | - Andrea Vallase
- Department of Environmental and Prevention Sciences, University of Ferrara, 44121 Ferrara, Italy
| | - Massimo Iorizzo
- Department of Horticultural Science, Plants for Human Health Institute, NC Research Campus, NC State University, Kannapolis, NC 28081, USA
| | - Alessandra Pecorelli
- Department of Food Bioprocessing & Nutrition Sciences, Plants for Human Health Institute, NC Research Campus, NC State University, Kannapolis, NC 28081, USA
- Department of Environmental and Prevention Sciences, University of Ferrara, 44121 Ferrara, Italy
| | - Mary Ann Lila
- Department of Food Bioprocessing & Nutrition Sciences, Plants for Human Health Institute, NC Research Campus, NC State University, Kannapolis, NC 28081, USA
| | - Giuseppe Valacchi
- Department of Animal Science, Plants for Human Health Institute, NC Research Campus, NC State University, Kannapolis, NC 28081, USA.
- Department of Environmental and Prevention Sciences, University of Ferrara, 44121 Ferrara, Italy
- Kyung Hee University, Department of Food and Nutrition, Seoul, South Korea
| |
Collapse
|
12
|
Banerjee SK, Thurlow LR, Kannan K, Richardson AR. Glucose transporter 1 is essential for the resolution of methicillin-resistant S. aureus skin and soft tissue infections. Cell Rep 2024; 43:114486. [PMID: 38990718 PMCID: PMC11323221 DOI: 10.1016/j.celrep.2024.114486] [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: 05/03/2024] [Revised: 06/03/2024] [Accepted: 06/25/2024] [Indexed: 07/13/2024] Open
Abstract
Skin/soft tissue infections (SSTIs) caused by methicillin-resistant Staphylococcus aureus (MRSA) pose a major healthcare burden. Distinct inflammatory and resolution phases comprise the host immune response to SSTIs. Resolution is a myeloid PPARγ-dependent anti-inflammatory phase that is essential for the clearance of MRSA. However, the signals activating PPARγ to induce resolution remain unknown. Here, we demonstrate that myeloid glucose transporter 1 (GLUT-1) is essential for the onset of resolution. MRSA-challenged macrophages are unsuccessful in generating an oxidative burst or immune radicals in the absence of GLUT-1 due to a reduction in the cellular NADPH pool. This translates in vivo as a significant reduction in lipid peroxidation products required for the activation of PPARγ in MRSA-infected mice lacking myeloid GLUT-1. Chemical induction of PPARγ during infection circumvents this GLUT-1 requirement and improves resolution. Thus, GLUT-1-dependent oxidative burst is essential for the activation of PPARγ and subsequent resolution of SSTIs.
Collapse
Affiliation(s)
- Srijon K Banerjee
- Department of Microbiology and Molecular Genetics, School of Medicine, University of Pittsburgh, 450 Technology Drive, Pittsburgh, PA 15219, USA
| | - Lance R Thurlow
- Adams School of Dentistry, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599-7450, USA
| | - Kartik Kannan
- Department of Microbiology and Molecular Genetics, School of Medicine, University of Pittsburgh, 450 Technology Drive, Pittsburgh, PA 15219, USA
| | - Anthony R Richardson
- Department of Microbiology and Molecular Genetics, School of Medicine, University of Pittsburgh, 450 Technology Drive, Pittsburgh, PA 15219, USA.
| |
Collapse
|
13
|
Zhang J, Chang J, Chen V, Beg MA, Huang W, Vick L, Wang Y, Zhang H, Yttre E, Gupta A, Castleberry M, Zhang Z, Dai W, Song S, Zhu J, Yang M, Brown AK, Xu Z, Ma YQ, Smith BC, Zielonka J, Traylor JG, Dhaou CB, Orr AW, Cui W, Zheng Z, Chen Y. Oxidized LDL regulates efferocytosis through the CD36-PKM2-mtROS pathway. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.09.07.556574. [PMID: 39071358 PMCID: PMC11275753 DOI: 10.1101/2023.09.07.556574] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/30/2024]
Abstract
Macrophage efferocytosis, the process by which phagocytes engulf and remove apoptotic cells (ACs), plays a critical role in maintaining tissue homeostasis. Efficient efferocytosis prevents secondary necrosis, mitigates chronic inflammation, and impedes atherosclerosis progression. However, the regulatory mechanisms of efferocytosis under atherogenic conditions remain poorly understood. We previously demonstrated that oxidized LDL (oxLDL), an atherogenic lipoprotein, induces mitochondrial reactive oxygen species (mtROS) in macrophages via CD36. In this study, we demonstrate that macrophage mtROS facilitate continual efferocytosis through a positive feedback mechanism. However, oxLDL disrupts continual efferocytosis by dysregulating the internalization of ACs. This disruption is mediated by an overproduction of mtROS. Mechanistically, oxLDL/CD36 signaling promotes the translocation of cytosolic PKM2 to mitochondria, facilitated by the chaperone GRP75. Mitochondrial PKM2 then binds to Complex III of the electron transport chain, inducing mtROS production. This study elucidates a novel regulatory mechanism of efferocytosis in atherosclerosis, providing potential therapeutic targets for intervention. SUMMARY Macrophages clear apoptotic cells through a process called efferocytosis, which involves mitochondrial ROS. However, the atherogenic oxidized LDL overstimulates mitochondrial ROS via the CD36-PKM2 pathway, disrupting continual efferocytosis. This finding elucidates a novel molecular mechanism that explains defects in efferocytosis, driving atherosclerosis progression.
Collapse
|
14
|
Primavesi F, Senoner T, Schindler S, Nikolajevic A, Di Fazio P, Csukovich G, Eller S, Neumayer B, Anliker M, Braunwarth E, Oberhuber R, Resch T, Maglione M, Cardini B, Niederwieser T, Gasteiger S, Klieser E, Tilg H, Schneeberger S, Neureiter D, Öfner D, Troppmair J, Stättner S. The Interplay between Perioperative Oxidative Stress and Hepatic Dysfunction after Human Liver Resection: A Prospective Observational Pilot Study. Antioxidants (Basel) 2024; 13:590. [PMID: 38790695 PMCID: PMC11118143 DOI: 10.3390/antiox13050590] [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: 04/05/2024] [Revised: 05/02/2024] [Accepted: 05/07/2024] [Indexed: 05/26/2024] Open
Abstract
Post-hepatectomy liver failure (PHLF) remains the major contributor to death after liver resection. Oxidative stress is associated with postoperative complications, but its impact on liver function is unclear. This first in-human, prospective, single-center, observational pilot study evaluated perioperative oxidative stress and PHLF according to the ISGLS (International Study Group for Liver Surgery). Serum 8-isoprostane, 4-hydroxynonenal (4-HNE), total antioxidative capacity, vitamins A and E, and intraoperative, sequential hepatic tissue 4-HNE and UCP2 (uncoupling protein 2) immunohistochemistry (IHC) were assessed. The interaction with known risk factors for PHLF and the predictive potential of oxidative stress markers were analyzed. Overall, 52 patients were included (69.2% major liver resection). Thirteen patients (25%) experienced PHLF, a major factor for 90-day mortality (23% vs. 0%; p = 0.013). Post-resection, pro-oxidative 8-isoprostane significantly increased (p = 0.038), while 4-HNE declined immediately (p < 0.001). Antioxidative markers showed patterns of consumption starting post-resection (p < 0.001). Liver tissue oxidative stress increased stepwise from biopsies taken after laparotomy to post-resection in situ liver and resection specimens (all p < 0.001). Cholangiocarcinoma patients demonstrated significantly higher serum and tissue oxidative stress levels at various timepoints, with consistently higher preoperative values in advanced tumor stages. Combining intraoperative, post-resection 4-HNE serum levels and in situ IHC early predicted PHLF with an AUC of 0.855 (63.6% vs. 0%; p < 0.001). This was also associated with grade B/C PHLF (36.4% vs. 0%; p = 0.021) and 90-day mortality (18.2% vs. 0%; p = 0.036). In conclusion, distinct patterns of perioperative oxidative stress levels occur in patients with liver dysfunction. Combining intraoperative serum and liver tissue markers predicts subsequent PHLF. Cholangiocarcinoma patients demonstrated pronounced systemic and hepatic oxidative stress, with increasing levels in advanced tumor stages, thus representing a worthwhile target for future exploratory and therapeutic studies.
Collapse
Affiliation(s)
- Florian Primavesi
- Department of Visceral, Transplant and Thoracic Surgery, Medical University of Innsbruck, 6020 Innsbruck, Austria; (S.S.); (E.B.); (R.O.); (T.R.); (M.M.); (B.C.); (S.G.); (S.S.); (D.Ö.)
- Daniel Swarovski Research Laboratory, Department of Visceral, Transplant and Thoracic Surgery, Medical University of Innsbruck, 6020 Innsbruck, Austria; (A.N.); (S.E.); (J.T.)
- Department of General, Visceral and Vascular Surgery, Salzkammergutklinikum, 4840 Vöcklabruck, Austria;
| | - Thomas Senoner
- Department of Anaesthesiology and Intensive Care Medicine, Medical University of Innsbruck, 6020 Innsbruck, Austria;
| | - Sophie Schindler
- Department of Visceral, Transplant and Thoracic Surgery, Medical University of Innsbruck, 6020 Innsbruck, Austria; (S.S.); (E.B.); (R.O.); (T.R.); (M.M.); (B.C.); (S.G.); (S.S.); (D.Ö.)
| | - Aleksandar Nikolajevic
- Daniel Swarovski Research Laboratory, Department of Visceral, Transplant and Thoracic Surgery, Medical University of Innsbruck, 6020 Innsbruck, Austria; (A.N.); (S.E.); (J.T.)
| | - Pietro Di Fazio
- Department of Visceral, Thoracic and Vascular Surgery, Philipps-Universität Marburg, 35043 Marburg, Germany;
| | - Georg Csukovich
- Daniel Swarovski Research Laboratory, Department of Visceral, Transplant and Thoracic Surgery, Medical University of Innsbruck, 6020 Innsbruck, Austria; (A.N.); (S.E.); (J.T.)
- Small Animal Internal Medicine, Vetmeduni, 1210 Vienna, Austria
| | - Silvia Eller
- Daniel Swarovski Research Laboratory, Department of Visceral, Transplant and Thoracic Surgery, Medical University of Innsbruck, 6020 Innsbruck, Austria; (A.N.); (S.E.); (J.T.)
| | - Bettina Neumayer
- Institute of Pathology, Paracelsus Medical University/University Hospital Salzburg (SALK), 5020 Salzburg, Austria; (B.N.); (E.K.); (D.N.)
| | - Markus Anliker
- Central Institute of Medical and Chemical Laboratory Diagnostics, Medical University of Innsbruck, 6020 Innsbruck, Austria;
| | - Eva Braunwarth
- Department of Visceral, Transplant and Thoracic Surgery, Medical University of Innsbruck, 6020 Innsbruck, Austria; (S.S.); (E.B.); (R.O.); (T.R.); (M.M.); (B.C.); (S.G.); (S.S.); (D.Ö.)
| | - Rupert Oberhuber
- Department of Visceral, Transplant and Thoracic Surgery, Medical University of Innsbruck, 6020 Innsbruck, Austria; (S.S.); (E.B.); (R.O.); (T.R.); (M.M.); (B.C.); (S.G.); (S.S.); (D.Ö.)
| | - Thomas Resch
- Department of Visceral, Transplant and Thoracic Surgery, Medical University of Innsbruck, 6020 Innsbruck, Austria; (S.S.); (E.B.); (R.O.); (T.R.); (M.M.); (B.C.); (S.G.); (S.S.); (D.Ö.)
| | - Manuel Maglione
- Department of Visceral, Transplant and Thoracic Surgery, Medical University of Innsbruck, 6020 Innsbruck, Austria; (S.S.); (E.B.); (R.O.); (T.R.); (M.M.); (B.C.); (S.G.); (S.S.); (D.Ö.)
- Daniel Swarovski Research Laboratory, Department of Visceral, Transplant and Thoracic Surgery, Medical University of Innsbruck, 6020 Innsbruck, Austria; (A.N.); (S.E.); (J.T.)
| | - Benno Cardini
- Department of Visceral, Transplant and Thoracic Surgery, Medical University of Innsbruck, 6020 Innsbruck, Austria; (S.S.); (E.B.); (R.O.); (T.R.); (M.M.); (B.C.); (S.G.); (S.S.); (D.Ö.)
| | - Thomas Niederwieser
- Department of Visceral, Transplant and Thoracic Surgery, Medical University of Innsbruck, 6020 Innsbruck, Austria; (S.S.); (E.B.); (R.O.); (T.R.); (M.M.); (B.C.); (S.G.); (S.S.); (D.Ö.)
| | - Silvia Gasteiger
- Department of Visceral, Transplant and Thoracic Surgery, Medical University of Innsbruck, 6020 Innsbruck, Austria; (S.S.); (E.B.); (R.O.); (T.R.); (M.M.); (B.C.); (S.G.); (S.S.); (D.Ö.)
| | - Eckhard Klieser
- Institute of Pathology, Paracelsus Medical University/University Hospital Salzburg (SALK), 5020 Salzburg, Austria; (B.N.); (E.K.); (D.N.)
| | - Herbert Tilg
- Department of Internal Medicine I, Gastroenterology, Hepatology, Endocrinology and Metabolism, Medical University of Innsbruck, 6020 Innsbruck, Austria;
| | - Stefan Schneeberger
- Department of Visceral, Transplant and Thoracic Surgery, Medical University of Innsbruck, 6020 Innsbruck, Austria; (S.S.); (E.B.); (R.O.); (T.R.); (M.M.); (B.C.); (S.G.); (S.S.); (D.Ö.)
| | - Daniel Neureiter
- Institute of Pathology, Paracelsus Medical University/University Hospital Salzburg (SALK), 5020 Salzburg, Austria; (B.N.); (E.K.); (D.N.)
| | - Dietmar Öfner
- Department of Visceral, Transplant and Thoracic Surgery, Medical University of Innsbruck, 6020 Innsbruck, Austria; (S.S.); (E.B.); (R.O.); (T.R.); (M.M.); (B.C.); (S.G.); (S.S.); (D.Ö.)
- Daniel Swarovski Research Laboratory, Department of Visceral, Transplant and Thoracic Surgery, Medical University of Innsbruck, 6020 Innsbruck, Austria; (A.N.); (S.E.); (J.T.)
| | - Jakob Troppmair
- Daniel Swarovski Research Laboratory, Department of Visceral, Transplant and Thoracic Surgery, Medical University of Innsbruck, 6020 Innsbruck, Austria; (A.N.); (S.E.); (J.T.)
| | - Stefan Stättner
- Department of General, Visceral and Vascular Surgery, Salzkammergutklinikum, 4840 Vöcklabruck, Austria;
| |
Collapse
|
15
|
Mansouri E, Asghari S, Nikooei P, Yaseri M, Vasheghani-Farahani A, Hosseinzadeh-Attar MJ. Effects of virgin coconut oil consumption on serum brain-derived neurotrophic factor levels and oxidative stress biomarkers in adults with metabolic syndrome: a randomized clinical trial. Nutr Neurosci 2024; 27:487-498. [PMID: 37409587 DOI: 10.1080/1028415x.2023.2223390] [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: 07/07/2023]
Abstract
BACKGROUND AND AIM Metabolic syndrome is associated with health conditions and neurological disorders. Brain-derived neurotrophic factor (BDNF) plays a protective role on the nervous system. Decreased levels of BDNF have been shown in MetS and neurodegenerative diseases. There is promising evidence regarding the anti-inflammatory antioxidant, and neuroprotective properties of virgin coconut oil (VCO). The aim of this study was to evaluate the effects of VCO consumption on serum BDNF levels, oxidative stress status, and insulin resistance in adults with MetS. METHODS This randomized controlled clinical trial was conducted on 48 adults with MetS aged 20-50 years. The intervention group received 30 ml of VCO daily to substitute the same amounts of oil in their usual diet. The control group continued their usual diet. Serum BDNF levels, total antioxidant capacity (TAC), malondialdehyde (MDA) as well as HOMA-IR and QUICKI index were measured after four weeks of intervention. RESULTS VCO consumption significantly reduced serum levels of MDA (p = .01), fasting insulin (p < .01) and HOMA-IR index (p < .01) and increased serum TAC (p < .01) and QUICKI index (p = .01) compared to the control group. Serum BDNF levels increased significantly in VCO group compared to the baseline (p = .02); however, this change was not significant when compared to the control group (p = .07). CONCLUSION VCO consumption improved oxidative stress status and insulin resistance and had a promising effect on BDNF levels in adults with MetS. Further studies are needed to understand the long-term effects of VCO consumption.
Collapse
Affiliation(s)
- Elahe Mansouri
- Department of Clinical Nutrition, Faculty of Nutritional Sciences and Dietetics, Tehran University of Medical Sciences, Tehran, Iran
| | - Somayyeh Asghari
- Department of Clinical Nutrition, Faculty of Nutritional Sciences and Dietetics, Tehran University of Medical Sciences, Tehran, Iran
| | - Parinaz Nikooei
- Department of Clinical Nutrition, Faculty of Nutritional Sciences and Dietetics, Tehran University of Medical Sciences, Tehran, Iran
| | - Mehdi Yaseri
- Department of Epidemiology and Biostatistics, Tehran University of Medical Sciences, Tehran, Iran
| | - Ali Vasheghani-Farahani
- Cardiac Primary Prevention Research Center (CPPRC), Tehran Heart Center, Tehran University of Medical Sciences, Tehran, Iran
- Department of Clinical Cardiac Electrophysiology, Tehran Heart Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohammad Javad Hosseinzadeh-Attar
- Department of Clinical Nutrition, Faculty of Nutritional Sciences and Dietetics, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
16
|
Morel L, Scindia Y. Functional consequence of Iron dyshomeostasis and ferroptosis in systemic lupus erythematosus and lupus nephritis. Clin Immunol 2024; 262:110181. [PMID: 38458303 DOI: 10.1016/j.clim.2024.110181] [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: 01/06/2024] [Accepted: 03/04/2024] [Indexed: 03/10/2024]
Abstract
Systemic lupus erythematosus (SLE) and its renal manifestation Lupus nephritis (LN) are characterized by a dysregulated immune system, autoantibodies, and injury to the renal parenchyma. Iron accumulation and ferroptosis in the immune effectors and renal tubules are recently identified pathological features in SLE and LN. Ferroptosis is an iron dependent non-apoptotic form of regulated cell death and ferroptosis inhibitors have improved disease outcomes in murine models of SLE, identifying it as a novel druggable target. In this review, we discuss novel mechanisms by which iron accumulation and ferroptosis perpetuate immune cell mediated pathology in SLE/LN. We highlight intra-renal dysregulation of iron metabolism and ferroptosis as an underlying pathogenic mechanism of renal tubular injury. The basic concepts of iron biology and ferroptosis are also discussed to expose the links between iron, cell metabolism and ferroptosis, that identify intracellular pro-ferroptotic enzymes and their protein conjugates as potential targets to improve SLE/LN outcomes.
Collapse
Affiliation(s)
- Laurence Morel
- Department of Microbiology, Immunology, and Molecular Genetics, UT Health San Antonio, San Antonio, TX, USA
| | - Yogesh Scindia
- Department of Medicine, University of Florida, Gainesville, FL, USA.
| |
Collapse
|
17
|
Rossetto IMU, Santos FR, da Silva HM, Minatel E, Mesquitta M, Salvador MJ, Montico F, Cagnon VHA. Tempol effect on oxidative and mitochondrial markers in preclinical models for prostate cancer. Toxicol Res (Camb) 2024; 13:tfae056. [PMID: 38623092 PMCID: PMC11015989 DOI: 10.1093/toxres/tfae056] [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: 11/09/2023] [Revised: 03/04/2024] [Accepted: 04/04/2024] [Indexed: 04/17/2024] Open
Abstract
Background Tempol is a redox-cycling nitroxide considered a potent antioxidant. The present study investigated the tempol effects on oxidative stress and mitochondrial markers on prostate cancer (PCa). Methods PC-3 and LnCaP cells were exposed to tempol. Cell viability test, western blot and Amplex Red analyses were performed. In vivo, five experimental groups evaluated tempol effects in the early (CT12 and TPL12 groups) and late stages (CT20, TPL20-I, and TLP20-II) of PCa development. The TPL groups were treated with 50 or 100 mg/kg tempol doses. Control groups received water as the vehicle. The ventral lobe of the prostate and the blood were collected and submitted to western blotting or enzymatic activity analyses. Results In vitro, tempol decreased cell viability and differentially altered the H2O2 content for PC-3 and LNCaP. Tempol increased SOD2 levels in both cell lines and did not alter Catalase protein levels. In vivo, tempol increased SOD2 levels in the early stage and did not change Catalase levels in the different PCa stages. Systemically, tempol decreased SOD2 levels in the late-stage and improved redox status in the early and late stages, which was confirmed by reduced LDH in tempol groups. Alterations on energetic metabolism and oxidative phosphorylation were observed in TRAMP model. Conclusion Tempol can be considered a beneficial therapy for PCa treatment considering its antioxidant and low toxicity properties, however the PCa progression must be evaluated to get successful therapy.
Collapse
Affiliation(s)
- Isabela Maria Urra Rossetto
- Department of Structural and Functional Biology, University of Campinas (UNICAMP), 255 Monteiro Lobato St., Campinas, SP 13083862, Brazil
| | - Felipe Rabelo Santos
- Department of Structural and Functional Biology, University of Campinas (UNICAMP), 255 Monteiro Lobato St., Campinas, SP 13083862, Brazil
| | - Heloina Mariano da Silva
- Department of Structural and Functional Biology, University of Campinas (UNICAMP), 255 Monteiro Lobato St., Campinas, SP 13083862, Brazil
| | - Elaine Minatel
- Department of Structural and Functional Biology, University of Campinas (UNICAMP), 255 Monteiro Lobato St., Campinas, SP 13083862, Brazil
| | - Mariana Mesquitta
- Department of Plant Biology, University of Campinas (UNICAMP), 255 Monteiro Lobato St., Campinas, SP 13083862, Brazil
| | - Marcos José Salvador
- Department of Plant Biology, University of Campinas (UNICAMP), 255 Monteiro Lobato St., Campinas, SP 13083862, Brazil
| | - Fábio Montico
- Department of Structural and Functional Biology, University of Campinas (UNICAMP), 255 Monteiro Lobato St., Campinas, SP 13083862, Brazil
| | - Valéria Helena Alves Cagnon
- Department of Structural and Functional Biology, University of Campinas (UNICAMP), 255 Monteiro Lobato St., Campinas, SP 13083862, Brazil
| |
Collapse
|
18
|
Wrońska A, Kieżun J, Kmieć Z. High-Dose Fenofibrate Stimulates Multiple Cellular Stress Pathways in the Kidney of Old Rats. Int J Mol Sci 2024; 25:3038. [PMID: 38474282 DOI: 10.3390/ijms25053038] [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: 01/24/2024] [Revised: 03/01/2024] [Accepted: 03/04/2024] [Indexed: 03/14/2024] Open
Abstract
We investigated the age-related effects of the lipid-lowering drug fenofibrate on renal stress-associated effectors. Young and old rats were fed standard chow with 0.1% or 0.5% fenofibrate. The kidney cortex tissue structure showed typical aging-related changes. In old rats, 0.1% fenofibrate reduced the thickening of basement membranes, but 0.5% fenofibrate exacerbated interstitial fibrosis. The PCR array for stress and toxicity-related targets showed that 0.1% fenofibrate mildly downregulated, whereas 0.5% upregulated multiple genes. In young rats, 0.1% fenofibrate increased some antioxidant genes' expression and decreased the immunoreactivity of oxidative stress marker 4-HNE. However, the activation of cellular antioxidant defenses was impaired in old rats. Fenofibrate modulated the expression of factors involved in hypoxia and osmotic stress signaling similarly in both age groups. Inflammatory response genes were variably modulated in the young rats, whereas old animals presented elevated expression of proinflammatory genes and TNFα immunoreactivity after 0.5% fenofibrate. In old rats, 0.1% fenofibrate more prominently than in young animals induced phospho-AMPK and PGC1α levels, and upregulated fatty acid oxidation genes. Our results show divergent effects of fenofibrate in young and old rat kidneys. The activation of multiple stress-associated effectors by high-dose fenofibrate in the aged kidney warrants caution when applying fenofibrate therapy to the elderly.
Collapse
Affiliation(s)
- Agata Wrońska
- Department of Histology, Faculty of Medicine, Medical University of Gdańsk, 80-210 Gdańsk, Poland
| | - Jacek Kieżun
- Department of Human Histology and Embryology, School of Medicine, University of Warmia and Mazury in Olsztyn, 10-082 Olsztyn, Poland
| | - Zbigniew Kmieć
- Department of Histology, Faculty of Medicine, Medical University of Gdańsk, 80-210 Gdańsk, Poland
| |
Collapse
|
19
|
Tsoneva E, Dimitrova P, Metodiev M, Shivarov V, Vasileva-Slaveva M, Yordanov A, Kostov S. Utility of expression of 4-hydroxynonenal tested by immunohistochemistry for cervical cancer. PRZEGLAD MENOPAUZALNY = MENOPAUSE REVIEW 2024; 23:6-13. [PMID: 38690070 PMCID: PMC11056727 DOI: 10.5114/pm.2024.136356] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Accepted: 11/17/2023] [Indexed: 05/02/2024]
Abstract
Introduction Cervical cancer (CC) is a leading cause of mortality in women around the world, with the highest incidence rate still being in developing countries. The most common aetiological factor is infection with high-risk human papilloma virus viral strains. Oxidative stress through generation of reactive oxygen species leads to lipid peroxidation and DNA damage. Studies show that reactive lipid electrophiles such as 4-hydroxynonenal (4-HNE) produced in the process play an important role in cancer signalling pathways and are a good biomarker for oxidative stress. We aim to investigate the prognostic role of 4-HNE as a biomarker for oxidative stress in patients in early and advanced stages of CC measured by immunohistochemistry. Material and methods This is a retrospective study of 69 patients treated at our Department of Oncogynaecology. Paraffin embedded tumour tissues were immunohistochemically tested for the levels of expression of 4-HNE. The results for H-score, Allred score, and combined score were investigated for association with tumour size, lymph node status, andInternational Federation of Gynaecology and Obstetrics stage. Results 4-hydroxynonenal showed higher expression in more advanced stages of CC and in cases with involved lymph nodes. Tumour size was not associated with the levels of 4-HNE. Conclusions To best of our knowledge, this is the first study to use immunohistochemistry to examine the expression of 4-HNE as a prognostic factor in CC. The 3 score systems showed similar results. The pattern of 4-HNE histological appearance is dependent on the histological origin of cancer and is not universal.
Collapse
Affiliation(s)
| | - Polina Dimitrova
- Department of Pathology, Medical University Pleven, Pleven, Bulgaria
| | | | | | | | - Angel Yordanov
- Department of Gynaecological Oncology, Medical University Pleven, Pleven, Bulgaria
| | - Stoyan Kostov
- Department of Gynaecology, Medical University Varna “Prof. Dr. Paraskev Stoyanov”, Varna, Bulgaria
| |
Collapse
|
20
|
Takashima S, Tokiya M, Izui K, Miyamoto H, Matsumoto A. Asian flush is a potential protective factor against COVID-19: a web-based retrospective survey in Japan. Environ Health Prev Med 2024; 29:14. [PMID: 38462476 PMCID: PMC10937249 DOI: 10.1265/ehpm.23-00361] [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: 12/14/2023] [Accepted: 02/10/2024] [Indexed: 03/12/2024] Open
Abstract
BACKGROUND Coronavirus disease 2019 (COVID-19), first reported in December 2019, spread worldwide in a short period, resulting in numerous cases and associated deaths; however, the toll was relatively low in East Asia. A genetic polymorphism unique to East Asians, Aldehyde dehydrogenase 2 rs671, has been reported to confer protection against infections. METHOD We retrospectively investigated the association between the surrogate marker of the rs671 variant, the skin flushing phenomenon after alcohol consumption, and the timing of COVID-19 incidence using a web-based survey tool to test any protective effects of rs671 against COVID-19. RESULTS A total of 807 valid responses were received from 362 non-flushers and 445 flushers. During the 42 months, from 12/1/2019 to 5/31/2023, 40.6% of non-flushers and 35.7% of flushers experienced COVID-19. Flushers tended to have a later onset (Spearman's partial rank correlation test, p = 0.057, adjusted for sex and age). Similarly, 2.5% of non-flushers and 0.5% of flushers were hospitalized because of COVID-19. Survival analysis estimated lower risks of COVID-19 and associated hospitalization among flushers (p = 0.03 and <0.01, respectively; generalized Wilcoxon test). With the Cox proportional hazards model covering 21 months till 8/31/2021, when approximately half of the Japanese population had received two doses of COVID-19 vaccine, the hazard ratio (95% confidence interval) of COVID-19 incidence was estimated to be 0.21 (0.10-0.46) for flusher versus non-flusher, with adjustment for sex, age, steroid use, and area of residence. CONCLUSIONS Our study suggests an association between the flushing phenomenon after drinking and a decreased risk of COVID-19 morbidity and hospitalization, suggesting that the rs671 variant is a protective factor. This study provides valuable information for infection control and helps understand the unique constitutional diversity of East Asians.
Collapse
Affiliation(s)
- Satoshi Takashima
- Department of Social and Environmental Medicine, Saga University, 5-1-1 Nabeshima, Saga 849-8501, Japan
- Plant Products Safety Division, Food Safety and Consumer Affairs Bureau, Ministry of Agriculture, Forestry and Fisheries, 1-2-1 Kasumigaseki, Chiyodaku, Tokyo 100-8950, Japan
| | - Mikiko Tokiya
- Department of Social and Environmental Medicine, Saga University, 5-1-1 Nabeshima, Saga 849-8501, Japan
| | - Katsura Izui
- Graduate School of Biostudies, Kyoto University, Oiwake-cho, Kitashirakawa, Sakyo-ku, Kyoto 606-8502, Japan
| | - Hiroshi Miyamoto
- Department of Pathology and Microbiology, Faculty of Medicine, Saga University, 5-1-1 Nabeshima, Saga 849-8501, Japan
| | - Akiko Matsumoto
- Department of Social and Environmental Medicine, Saga University, 5-1-1 Nabeshima, Saga 849-8501, Japan
| |
Collapse
|
21
|
Chen S, Pang J, Huang R, You Y, Zhang H, Xue H, Chen X. Associations of Macronutrients Intake With MRI-determined Hepatic Fat Content, Hepatic Fibroinflammation, and NAFLD. J Clin Endocrinol Metab 2023; 108:e1660-e1669. [PMID: 37290038 DOI: 10.1210/clinem/dgad346] [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: 05/05/2023] [Revised: 05/31/2023] [Accepted: 06/05/2023] [Indexed: 06/10/2023]
Abstract
CONTEXT A healthy lifestyle is the cornerstone of management in nonalcoholic fatty liver disease (NAFLD). However, the associations between dietary macronutrient composition and different aspects of NAFLD pathology are unclear and dietary recommendations for NAFLD are lacking. OBJECTIVE This work aimed to evaluate the associations of dietary macronutrient composition with hepatic steatosis, hepatic fibroinflammation, and NAFLD. METHODS In this cross-sectional study, a total of 12 620 UK Biobank participants who completed both the dietary questionnaire and magnetic resonance imaging (MRI) examination were included in this study. Dietary consumption of macronutrient was self-reported and calculated. MRI-determined hepatic fat content, fibroinflammation, and NAFLD were estimated. RESULTS First, we found that saturated fatty acid (SFA) intake was associated with higher hepatic steatosis, fibroinflammation, and NAFLD prevalence. In contrast, higher fiber or protein intake was reversely correlated with hepatic steatosis and fibroinflammation. Interestingly, starch or sugar intake was significantly associated with hepatic fibroinflammation, whereas monounsaturated fatty acid (MUFA) intake was negatively correlated with hepatic fibroinflammation. Isocaloric analysis revealed that replacing SFA with sugar, fiber, or protein was significantly associated with a reduction in hepatic steatosis, while replacing starch, sugar, or SFA with protein or MUFA was significantly correlated with a decrease in hepatic fibroinflammation. CONCLUSION Overall, our results demonstrate that specific macronutrients are associated with different aspects of NAFLD, and specific dietary compositions should be recommended for distinct NAFLD-risk populations.
Collapse
Affiliation(s)
- Shen Chen
- Department of Toxicology, School of Public Health, Sun Yat-sen University, Guangzhou 510080, People's Republic of China
| | - Juan Pang
- Department of Nutrition, School of Public Health, Sun Yat-sen University, Guangzhou 510080, People's Republic of China
- Laboratory of Clinical Pharmacy and Adverse Drug Reaction, West China Hospital, Sichuan University, Chengdu 610041, People's Republic of China
| | - Rong Huang
- Medical Science and Technology Innovation Center, Jinan Central Hospital, Shandong First Medical University, Shandong 250013, People's Republic of China
| | - Yiran You
- Department of Nutrition, School of Public Health, Sun Yat-sen University, Guangzhou 510080, People's Republic of China
| | - Haoyang Zhang
- School of Data and Computer Science, Sun Yat-sen University, Guangzhou 510080, China
| | - Hongliang Xue
- Department of Nutrition, School of Public Health, Guangzhou Medical University, Guangzhou 510080, People's Republic of China
| | - Xu Chen
- Department of Nutrition, School of Public Health, Sun Yat-sen University, Guangzhou 510080, People's Republic of China
- Department of Molecular, Cellular, and Developmental Biology, University of Colorado, Boulder, CO 80302, USA
| |
Collapse
|
22
|
Georgiou-Siafis SK, Tsiftsoglou AS. The Key Role of GSH in Keeping the Redox Balance in Mammalian Cells: Mechanisms and Significance of GSH in Detoxification via Formation of Conjugates. Antioxidants (Basel) 2023; 12:1953. [PMID: 38001806 PMCID: PMC10669396 DOI: 10.3390/antiox12111953] [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: 09/30/2023] [Revised: 10/18/2023] [Accepted: 10/23/2023] [Indexed: 11/26/2023] Open
Abstract
Glutathione (GSH) is a ubiquitous tripeptide that is biosynthesized in situ at high concentrations (1-5 mM) and involved in the regulation of cellular homeostasis via multiple mechanisms. The main known action of GSH is its antioxidant capacity, which aids in maintaining the redox cycle of cells. To this end, GSH peroxidases contribute to the scavenging of various forms of ROS and RNS. A generally underestimated mechanism of action of GSH is its direct nucleophilic interaction with electrophilic compounds yielding thioether GSH S-conjugates. Many compounds, including xenobiotics (such as NAPQI, simvastatin, cisplatin, and barbital) and intrinsic compounds (such as menadione, leukotrienes, prostaglandins, and dopamine), form covalent adducts with GSH leading mainly to their detoxification. In the present article, we wish to present the key role and significance of GSH in cellular redox biology. This includes an update on the formation of GSH-S conjugates or GSH adducts with emphasis given to the mechanism of reaction, the dependence on GST (GSH S-transferase), where this conjugation occurs in tissues, and its significance. The uncovering of the GSH adducts' formation enhances our knowledge of the human metabolome. GSH-hematin adducts were recently shown to have been formed spontaneously in multiples isomers at hemolysates, leading to structural destabilization of the endogenous toxin, hematin (free heme), which is derived from the released hemoglobin. Moreover, hemin (the form of oxidized heme) has been found to act through the Kelch-like ECH associated protein 1 (Keap1)-nuclear factor erythroid 2-related factor-2 (Nrf2) signaling pathway as an epigenetic modulator of GSH metabolism. Last but not least, the implications of the genetic defects in GSH metabolism, recorded in hemolytic syndromes, cancer and other pathologies, are presented and discussed under the framework of conceptualizing that GSH S-conjugates could be regarded as signatures of the cellular metabolism in the diseased state.
Collapse
Affiliation(s)
| | - Asterios S. Tsiftsoglou
- Laboratory of Pharmacology, Department of Pharmaceutical Sciences, School of Health Sciences, Aristotle University of Thessaloniki (AUTh), 54124 Thessaloniki, Greece;
| |
Collapse
|
23
|
Reddy VP. Oxidative Stress in Health and Disease. Biomedicines 2023; 11:2925. [PMID: 38001926 PMCID: PMC10669448 DOI: 10.3390/biomedicines11112925] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Revised: 10/25/2023] [Accepted: 10/26/2023] [Indexed: 11/26/2023] Open
Abstract
Oxidative stress, resulting from the excessive intracellular accumulation of reactive oxygen species (ROS), reactive nitrogen species (RNS), and other free radical species, contributes to the onset and progression of various diseases, including diabetes, obesity, diabetic nephropathy, diabetic neuropathy, and neurological diseases, such as Alzheimer's disease (AD), amyotrophic lateral sclerosis (ALS), and Parkinson's disease (PD). Oxidative stress is also implicated in cardiovascular disease and cancer. Exacerbated oxidative stress leads to the accelerated formation of advanced glycation end products (AGEs), a complex mixture of crosslinked proteins and protein modifications. Relatively high levels of AGEs are generated in diabetes, obesity, AD, and other I neurological diseases. AGEs such as Ne-carboxymethyllysine (CML) serve as markers for disease progression. AGEs, through interaction with receptors for advanced glycation end products (RAGE), initiate a cascade of deleterious signaling events to form inflammatory cytokines, and thereby further exacerbate oxidative stress in a vicious cycle. AGE inhibitors, AGE breakers, and RAGE inhibitors are therefore potential therapeutic agents for multiple diseases, including diabetes and AD. The complexity of the AGEs and the lack of well-established mechanisms for AGE formation are largely responsible for the lack of effective therapeutics targeting oxidative stress and AGE-related diseases. This review addresses the role of oxidative stress in the pathogenesis of AGE-related chronic diseases, including diabetes and neurological disorders, and recent progress in the development of therapeutics based on antioxidants, AGE breakers and RAGE inhibitors. Furthermore, this review outlines therapeutic strategies based on single-atom nanozymes that attenuate oxidative stress through the sequestering of reactive oxygen species (ROS) and reactive nitrogen species (RNS).
Collapse
Affiliation(s)
- V Prakash Reddy
- Department of Chemistry, Missouri University of Science and Technology, Rolla, MO 65409, USA
| |
Collapse
|
24
|
Oliva CA, Lira M, Jara C, Catenaccio A, Mariqueo TA, Lindsay CB, Bozinovic F, Cavieres G, Inestrosa NC, Tapia-Rojas C, Rivera DS. Long-term social isolation stress exacerbates sex-specific neurodegeneration markers in a natural model of Alzheimer's disease. Front Aging Neurosci 2023; 15:1250342. [PMID: 37810621 PMCID: PMC10557460 DOI: 10.3389/fnagi.2023.1250342] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Accepted: 08/30/2023] [Indexed: 10/10/2023] Open
Abstract
Social interactions have a significant impact on health in humans and animal models. Social isolation initiates a cascade of stress-related physiological disorders and stands as a significant risk factor for a wide spectrum of morbidity and mortality. Indeed, social isolation stress (SIS) is indicative of cognitive decline and risk to neurodegenerative conditions, including Alzheimer's disease (AD). This study aimed to evaluate the impact of chronic, long-term SIS on the propensity to develop hallmarks of AD in young degus (Octodon degus), a long-lived animal model that mimics sporadic AD naturally. We examined inflammatory factors, bioenergetic status, reactive oxygen species (ROS), oxidative stress, antioxidants, abnormal proteins, tau protein, and amyloid-β (Aβ) levels in the hippocampus of female and male degus that were socially isolated from post-natal and post-weaning until adulthood. Additionally, we explored the effect of re-socialization following chronic isolation on these protein profiles. Our results showed that SIS promotes a pro-inflammatory scenario more severe in males, a response that was partially mitigated by a period of re-socialization. In addition, ATP levels, ROS, and markers of oxidative stress are severely affected in female degus, where a period of re-socialization fails to restore them as it does in males. In females, these effects might be linked to antioxidant enzymes like catalase, which experience a decline across all SIS treatments without recovery during re-socialization. Although in males, a previous enzyme in antioxidant pathway diminishes in all treatments, catalase rebounds during re-socialization. Notably, males have less mature neurons after chronic isolation, whereas phosphorylated tau and all detectable forms of Aβ increased in both sexes, persisting even post re-socialization. Collectively, these findings suggest that long-term SIS may render males more susceptible to inflammatory states, while females are predisposed to oxidative states. In both scenarios, the accumulation of tau and Aβ proteins increase the individual susceptibility to early-onset neurodegenerative conditions such as AD.
Collapse
Affiliation(s)
- Carolina A. Oliva
- Centro para la Transversalización de Género en I+D+i+e, Vicerrectoría de Investigación y Doctorados, Universidad Autónoma de Chile, Santiago, Chile
| | - Matías Lira
- Laboratory of Neurobiology of Aging, Centro de Biología Celular y Biomedicina (CEBICEM), Facultad de Medicina y Ciencia, Universidad San Sebastián, Santiago, Chile
- Centro Científico y Tecnológico de Excelencia Ciencia & Vida, Santiago, Chile
| | - Claudia Jara
- Laboratory of Neurobiology of Aging, Centro de Biología Celular y Biomedicina (CEBICEM), Facultad de Medicina y Ciencia, Universidad San Sebastián, Santiago, Chile
| | - Alejandra Catenaccio
- Laboratory of Neurobiology of Aging, Centro de Biología Celular y Biomedicina (CEBICEM), Facultad de Medicina y Ciencia, Universidad San Sebastián, Santiago, Chile
- Centro Científico y Tecnológico de Excelencia Ciencia & Vida, Santiago, Chile
| | - Trinidad A. Mariqueo
- Centro de Investigaciones Médicas, Laboratorio de Neurofarmacología, Escuela de Medicina, Universidad de Talca, Talca, Chile
| | - Carolina B. Lindsay
- Laboratory of Neurosystems, Department of Neuroscience and Biomedical Neuroscience Institute, Faculty of Medicine, Universidad de Chile, Santiago, Chile
| | - Francisco Bozinovic
- Center of Applied Ecology and Sustainability (CAPES), Departamento de Ecología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Grisel Cavieres
- Center of Applied Ecology and Sustainability (CAPES), Departamento de Ecología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
- Departamento de Zoología, Facultad de Ciencias Naturales y Oceanográficas, Universidad de Concepción, Concepción, Chile
| | - Nibaldo C. Inestrosa
- Center of Aging and Regeneration UC (CARE-UC), Departamento de Biología Celular y Molecular, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
- Centro de Excelencia en Biomedicina de Magallanes (CEBIMA), Universidad de Magallanes, Punta Arenas, Chile
| | - Cheril Tapia-Rojas
- Laboratory of Neurobiology of Aging, Centro de Biología Celular y Biomedicina (CEBICEM), Facultad de Medicina y Ciencia, Universidad San Sebastián, Santiago, Chile
- Centro Científico y Tecnológico de Excelencia Ciencia & Vida, Santiago, Chile
| | - Daniela S. Rivera
- GEMA Center for Genomics, Ecology and Environment, Facultad de Ciencias, Ingeniería y Tecnología, Universidad Mayor, Santiago, Chile
| |
Collapse
|
25
|
Xanthis V, Mantso T, Dimtsi A, Pappa A, Fadouloglou VE. Human Aldehyde Dehydrogenases: A Superfamily of Similar Yet Different Proteins Highly Related to Cancer. Cancers (Basel) 2023; 15:4419. [PMID: 37686694 PMCID: PMC10650815 DOI: 10.3390/cancers15174419] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Revised: 08/30/2023] [Accepted: 09/01/2023] [Indexed: 09/10/2023] Open
Abstract
The superfamily of human aldehyde dehydrogenases (hALDHs) consists of 19 isoenzymes which are critical for several physiological and biosynthetic processes and play a major role in the organism's detoxification via the NAD(P) dependent oxidation of numerous endogenous and exogenous aldehyde substrates to their corresponding carboxylic acids. Over the last decades, ALDHs have been the subject of several studies as it was revealed that their differential expression patterns in various cancer types are associated either with carcinogenesis or promotion of cell survival. Here, we attempt to provide a thorough review of hALDHs' diverse functions and 3D structures with particular emphasis on their role in cancer pathology and resistance to chemotherapy. We are especially interested in findings regarding the association of structural features and their changes with effects on enzymes' functionalities. Moreover, we provide an updated outline of the hALDHs inhibitors utilized in experimental or clinical settings for cancer therapy. Overall, this review aims to provide a better understanding of the impact of ALDHs in cancer pathology and therapy from a structural perspective.
Collapse
Affiliation(s)
| | | | | | | | - Vasiliki E. Fadouloglou
- Department of Molecular Biology & Genetics, Democritus University of Thrace, 68100 Alexandroupolis, Greece
| |
Collapse
|
26
|
Tong Y, Wu Y, Ma J, Ikeda M, Ide T, Griffin CT, Ding XQ, Wang S. Comparative mechanistic study of RPE cell death induced by different oxidative stresses. Redox Biol 2023; 65:102840. [PMID: 37566944 PMCID: PMC10440584 DOI: 10.1016/j.redox.2023.102840] [Citation(s) in RCA: 15] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2023] [Revised: 07/31/2023] [Accepted: 08/04/2023] [Indexed: 08/13/2023] Open
Abstract
Oxidative stress is hypothesized to drive the progression of age-related macular degeneration (AMD). Retinal pigment epithelial (RPE) cell layer is important for supporting the function of retina and is particularly susceptible to oxidative stress-induced cell death. How RPE cells die in AMD, especially in geographic atrophy (GA), a late stage of dry AMD, is still controversial. The goal of this study is to compare the features and mechanisms of RPE cell death induced by different oxidative stresses, to identify potential universal therapeutic targets for GA. RPE cell death was induced both in vitro and ex vivo by 4-Hydroxynonenal (4-HNE), a major product of lipid peroxidation, sodium iodate (NaIO3) that has been widely used to model RPE cell death in dry AMD, a ferroptosis inducer RAS-selective lethal 3 (RSL3) or a necroptosis inducer shikonin. We found that RPE necroptosis and ferroptosis show common and distinct features. Common features include receptor-interacting protein kinase (RIPK)1/RIPK3 activation and lipid reactive oxygen species (ROS) accumulation, although lipid ROS accumulation is much milder during necroptosis. This supports cross talk between RPE ferroptosis and necroptosis pathways and is consistent with the rescue of RPE necroptosis and ferroptosis by RIPK1 inhibitor Necrostatin-1 (Nec-1) or in Ripk3-/- RPE explants. Distinct feature includes activated mixed lineage kinase domain like pseudokinase (MLKL) that is translocated to the cell membrane during necroptosis, which is not happening in ferroptosis. This is consistent with the failure to rescue RPE ferroptosis by MLKL inhibitor necrosulfonamide (NSA) or in Mlkl-/- RPE explants. Using this framework, we found that 4-HNE and NaIO3 induced RPE cell death likely through necroptosis based on the molecular features and the rescuing effect by multiple inhibitors. Our studies suggest that multiple markers and inhibitors are required to distinguish RPE necroptosis and ferroptosis, and that necroptosis inhibitor Nec-1 could be a potential therapeutic compound for GA since it inhibits RIPK1/RIPK3 activation and lipid ROS accumulation occurred in both necroptosis and ferroptosis pathways.
Collapse
Affiliation(s)
- Yao Tong
- Department of Cell and Molecular Biology, Tulane University, New Orleans, LA, 70118, USA
| | - Yinga Wu
- Department of Cell and Molecular Biology, Tulane University, New Orleans, LA, 70118, USA
| | - Jing Ma
- Department of Cell and Molecular Biology, Tulane University, New Orleans, LA, 70118, USA
| | - Masataka Ikeda
- Department of Cardiovascular Medicine, Faculty of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Tomomi Ide
- Department of Cardiovascular Medicine, Faculty of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Courtney T Griffin
- Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA; Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
| | - Xi-Qin Ding
- Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA
| | - Shusheng Wang
- Department of Cell and Molecular Biology, Tulane University, New Orleans, LA, 70118, USA; Department of Ophthalmology, Tulane University, New Orleans, LA, 70118, USA; Tulane Personalized Health Institute, Tulane University, New Orleans, LA, 70112, USA.
| |
Collapse
|
27
|
Berry SB, Espich S, Thuong NTT, Chang X, Dorajoo R, Khor CC, Heng CK, Yuan JM, Fox D, Anaya-Sanchez A, Tenney L, Chang CJ, Kotov DI, Vance RE, Dunstan SJ, Darwin KH, Stanley SA. Disruption of Aldehyde Dehydrogenase 2 protects against bacterial infection. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.08.24.554661. [PMID: 37662190 PMCID: PMC10473740 DOI: 10.1101/2023.08.24.554661] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/05/2023]
Abstract
The ALDH2*2 (rs671) allele is one of the most common genetic mutations in humans, yet the positive evolutionary selective pressure to maintain this mutation is unknown, despite its association with adverse health outcomes. ALDH2 is responsible for the detoxification of metabolically produced aldehydes, including lipid-peroxidation end products derived from inflammation. Here, we demonstrate that host-derived aldehydes 4-hydroxynonenal (4HNE), malondialdehyde (MDA), and formaldehyde (FA), all of which are metabolized by ALDH2, are directly toxic to the bacterial pathogens Mycobacterium tuberculosis and Francisella tularensis at physiological levels. We find that Aldh2 expression in macrophages is decreased upon immune stimulation, and that bone marrow-derived macrophages from Aldh2 -/- mice contain elevated aldehydes relative to wild-type mice. Macrophages deficient for Aldh2 exhibited enhanced control of Francisella infection. Finally , mice lacking Aldh2 demonstrated increased resistance to pulmonary infection by M. tuberculosis , including in a hypersusceptible model of tuberculosis, and were also resistant to Francisella infection. We hypothesize that the absence of ALDH2 contributes to the host's ability to control infection by pathogens such as M. tuberculosis and F. tularensis , and that host-derived aldehydes act as antimicrobial factors during intracellular bacterial infections. One sentence summary Aldehydes produced by host cells contribute to the control of bacterial infections.
Collapse
|
28
|
Wang Y, Anesi J, Maier MC, Myers MA, Oqueli E, Sobey CG, Drummond GR, Denton KM. Sympathetic Nervous System and Atherosclerosis. Int J Mol Sci 2023; 24:13132. [PMID: 37685939 PMCID: PMC10487841 DOI: 10.3390/ijms241713132] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2023] [Revised: 08/18/2023] [Accepted: 08/22/2023] [Indexed: 09/10/2023] Open
Abstract
Atherosclerosis is characterized by the narrowing of the arterial lumen due to subendothelial lipid accumulation, with hypercholesterolemia being a major risk factor. Despite the recent advances in effective lipid-lowering therapies, atherosclerosis remains the leading cause of mortality globally, highlighting the need for additional therapeutic strategies. Accumulating evidence suggests that the sympathetic nervous system plays an important role in atherosclerosis. In this article, we reviewed the sympathetic innervation in the vasculature, norepinephrine synthesis and metabolism, sympathetic activity measurement, and common signaling pathways of sympathetic activation. The focus of this paper was to review the effectiveness of pharmacological antagonists or agonists of adrenoceptors (α1, α2, β1, β2, and β3) and renal denervation on atherosclerosis. All five types of adrenoceptors are present in arterial blood vessels. α1 blockers inhibit atherosclerosis but increase the risk of heart failure while α2 agonism may protect against atherosclerosis and newer generations of β blockers and β3 agonists are promising therapies against atherosclerosis; however, new randomized controlled trials are warranted to investigate the effectiveness of these therapies in atherosclerosis inhibition and cardiovascular risk reduction in the future. The role of renal denervation in atherosclerosis inhibition in humans is yet to be established.
Collapse
Affiliation(s)
- Yutang Wang
- Discipline of Life Science, Institute of Innovation, Science and Sustainability, Federation University Australia, Ballarat, VIC 3350, Australia
| | - Jack Anesi
- Discipline of Life Science, Institute of Innovation, Science and Sustainability, Federation University Australia, Ballarat, VIC 3350, Australia
| | - Michelle C. Maier
- Discipline of Life Science, Institute of Innovation, Science and Sustainability, Federation University Australia, Ballarat, VIC 3350, Australia
| | - Mark A. Myers
- Discipline of Life Science, Institute of Innovation, Science and Sustainability, Federation University Australia, Ballarat, VIC 3350, Australia
| | - Ernesto Oqueli
- Cardiology Department, Grampians Health Ballarat, Ballarat, VIC 3350, Australia
- School of Medicine, Faculty of Health, Deakin University, Geelong, VIC 3216, Australia
| | - Christopher G. Sobey
- Centre for Cardiovascular Biology and Disease Research, Department of Microbiology, Anatomy, Physiology & Pharmacology, School of Agriculture, Biomedicine & Environment, La Trobe University, Melbourne, VIC 3086, Australia
| | - Grant R. Drummond
- Centre for Cardiovascular Biology and Disease Research, Department of Microbiology, Anatomy, Physiology & Pharmacology, School of Agriculture, Biomedicine & Environment, La Trobe University, Melbourne, VIC 3086, Australia
| | - Kate M. Denton
- Department of Physiology, Monash University, Melbourne, VIC 3800, Australia
- Cardiovascular Disease Program, Monash Biomedicine Discovery Institute, Monash University, Melbourne, VIC 3800, Australia
| |
Collapse
|
29
|
Orešić T, Bubanović S, Ramić S, Šarčević B, Čipak Gašparović A. Nuclear localization of NRF2 in stroma of HER2 positive and triple-negative breast cancer. Pathol Res Pract 2023; 248:154662. [PMID: 37421843 DOI: 10.1016/j.prp.2023.154662] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Revised: 06/28/2023] [Accepted: 06/30/2023] [Indexed: 07/10/2023]
Abstract
Breast cancer is one of the leading causes of cancer-related mortality in women. During tumor growth, periods of hypoxia are followed by reoxygenation due to neovascularisation leading to disturbed redox homeostasis. ROS (Reactive Oxygen Species) produced under hypoxia activate HIF1α. ROS can also activate the major antioxidant transcription factor NRF2, but also cause damage to biomolecules. Lipids are susceptible to peroxidation, as evidenced by the formation of reactive aldehydes, among which, HNE (4-hydroxynonenal) is the most studied one. Knowing that HIF1α (Hypoxia Inducing Factor 1α) is associated with breast cancer malignancy, we aimed to investigate its correlation with HNE and NRF2 (Nuclear factor erythroid 2-related factor 2). Our results show that HIF1α is activated in breast cancer, indicating an increase in ROS but not followed by HNE production. On the other hand, NRF2 was increased in all types of breast cancer suggesting that oxidative stress is present in these pathologies, but also supporting HIF1α. Interestingly, NRF2 was activated in HER2 positive and TNBC, indicating the role of stromal NRF2 in breast cancer malignancy.
Collapse
Affiliation(s)
- Tomislav Orešić
- University Hospital for Tumors, University Hospital Centre "Sestre milosrdnice", Ilica 197, HR-10000 Zagreb, Croatia.
| | - Sanda Bubanović
- University Hospital for Tumors, University Hospital Centre "Sestre milosrdnice", Ilica 197, HR-10000 Zagreb, Croatia.
| | - Snježana Ramić
- University Hospital for Tumors, University Hospital Centre "Sestre milosrdnice", Ilica 197, HR-10000 Zagreb, Croatia.
| | - Božena Šarčević
- University Hospital for Tumors, University Hospital Centre "Sestre milosrdnice", Ilica 197, HR-10000 Zagreb, Croatia.
| | | |
Collapse
|
30
|
Mercola J, D'Adamo CR. Linoleic Acid: A Narrative Review of the Effects of Increased Intake in the Standard American Diet and Associations with Chronic Disease. Nutrients 2023; 15:3129. [PMID: 37513547 PMCID: PMC10386285 DOI: 10.3390/nu15143129] [Citation(s) in RCA: 17] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Revised: 06/20/2023] [Accepted: 06/25/2023] [Indexed: 07/30/2023] Open
Abstract
The intake of linoleic acid (LA) has increased dramatically in the standard American diet. LA is generally promoted as supporting human health, but there exists controversy regarding whether the amount of LA currently consumed in the standard American diet supports human health. The goal of this narrative review is to explore the mechanisms that underlie the hypothesis that excessive LA intake may harm human health. While LA is considered to be an essential fatty acid and support health when consumed in modest amounts, an excessive intake of LA leads to the formation of oxidized linoleic acid metabolites (OXLAMs), impairments in mitochondrial function through suboptimal cardiolipin composition, and likely contributes to many chronic diseases that became an epidemic in the 20th century, and whose prevalence continues to increase. The standard American diet comprises 14 to 25 times more omega-6 fatty acids than omega-3 fatty acids, with the majority of omega-6 intake coming from LA. As LA consumption increases, the potential for OXLAM formation also increases. OXLAMs have been associated with various illnesses, including cardiovascular disease, cancer, and Alzheimer's disease, among others. Lowering dietary LA intake can help reduce the production and accumulation of OXLAMs implicated in chronic diseases. While there are other problematic components in the standard American diet, the half-life of LA is approximately two years, which means the damage can be far more persistent than other dietary factors, and the impact of reducing excessive LA intake takes time. Therefore, additional research-evaluating approaches to reduce OXLAM formation and cardiolipin derangements following LA consumption are warranted.
Collapse
Affiliation(s)
- Joseph Mercola
- Natural Health Partners, LLC, 125 SW 3rd Place, Cape Coral, FL 33991, USA
| | - Christopher R D'Adamo
- Department of Family and Community Medicine, Center for Integrative Medicine, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| |
Collapse
|
31
|
Vijayraghavan S, Saini N. Aldehyde-Associated Mutagenesis─Current State of Knowledge. Chem Res Toxicol 2023. [PMID: 37363863 DOI: 10.1021/acs.chemrestox.3c00045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/28/2023]
Abstract
Aldehydes are widespread in the environment, with multiple sources such as food and beverages, industrial effluents, cigarette smoke, and additives. The toxic effects of exposure to several aldehydes have been observed in numerous studies. At the molecular level, aldehydes damage DNA, cross-link DNA and proteins, lead to lipid peroxidation, and are associated with increased disease risk including cancer. People genetically predisposed to aldehyde sensitivity exhibit severe health outcomes. In various diseases such as Fanconi's anemia and Cockayne syndrome, loss of aldehyde-metabolizing pathways in conjunction with defects in DNA repair leads to widespread DNA damage. Importantly, aldehyde-associated mutagenicity is being explored in a growing number of studies, which could offer key insights into how they potentially contribute to tumorigenesis. Here, we review the genotoxic effects of various aldehydes, focusing particularly on the DNA adducts underlying the mutagenicity of environmentally derived aldehydes. We summarize the chemical structures of the aldehydes and their predominant DNA adducts, discuss various methodologies, in vitro and in vivo, commonly used in measuring aldehyde-associated mutagenesis, and highlight some recent studies looking at aldehyde-associated mutation signatures and spectra. We conclude the Review with a discussion on the challenges and future perspectives of investigating aldehyde-associated mutagenesis.
Collapse
Affiliation(s)
- Sriram Vijayraghavan
- Department of Biochemistry and Molecular Biology, Medical University of South Carolina, Charleston, South Carolina 29425, United States
| | - Natalie Saini
- Department of Biochemistry and Molecular Biology, Medical University of South Carolina, Charleston, South Carolina 29425, United States
| |
Collapse
|
32
|
Liu Z, Yuan J, Wen P, Guo X, Li K, Wang Y, Liu R, Guo Y, Li D. Effect of Lard or Plus Soybean Oil on Markers of Liver Function in Healthy Subjects: A Randomized Controlled-Feeding Trial. Foods 2023; 12:foods12091894. [PMID: 37174432 PMCID: PMC10178189 DOI: 10.3390/foods12091894] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Revised: 04/15/2023] [Accepted: 04/27/2023] [Indexed: 05/15/2023] Open
Abstract
Humans have consumed lard for thousands of years, but in recent decades, it has become much less popular because it is regarded as saturated fat. Animal studies showed that lard plus soybean oil (blend oil) was more advantageous for liver health than using either oil alone. This study aims to assess the effects of blend oil on liver function markers in healthy subjects. The 345 healthy subjects were randomized into 3 isoenergetic diet groups with different edible oils (30 g/day) (soybean oil, lard, and blend oil (50% lard and 50% soybean oil)) for 12 weeks. The reductions in both aspartate aminotransferase (AST) and alanine aminotransferase (ALT) were greater in the blend oil group than in the two other groups (p = 0.001 and <0.001 for the interaction between diet group and time, respectively). The reductions in AST and ALT in the blend oil group were more significant compared with those in the soybean oil group (p < 0.001) or lard group (p < 0.001). There were no significant differences in the other liver function markers between the groups. Thus, blend oil was beneficial for liver function markers such as AST and ALT compared with soybean oil and lard alone, which might help prevent non-alcoholic fatty liver disease in the healthy population.
Collapse
Affiliation(s)
- Zhiyuan Liu
- Institute of Nutrition & Health, Qingdao University, Qingdao 266071, China
| | - Jihong Yuan
- No. 2 Department of Nutrition, Chinese People's Liberation Army General Hospital, Beijing 100853, China
| | - Ping Wen
- Supply Department, Chinese People's Liberation Army General Hospital, Beijing 100853, China
| | - Xiaofei Guo
- Institute of Nutrition & Health, Qingdao University, Qingdao 266071, China
| | - Kelei Li
- Institute of Nutrition & Health, Qingdao University, Qingdao 266071, China
| | - Yinpeng Wang
- Institute of Nutrition & Health, Qingdao University, Qingdao 266071, China
| | - Ruirui Liu
- Institute of Nutrition & Health, Qingdao University, Qingdao 266071, China
| | - Yanjun Guo
- Institute of Nutrition & Health, Qingdao University, Qingdao 266071, China
| | - Duo Li
- Institute of Nutrition & Health, Qingdao University, Qingdao 266071, China
- Department of Food Science and Nutrition, Zhejiang University, Hangzhou 310058, China
- Department of Nutrition, Dietetics and Food, Monash University, Melbourne 3800, Australia
| |
Collapse
|
33
|
Do Q, Zhang R, Hooper G, Xu L. Differential Contributions of Distinct Free Radical Peroxidation Mechanisms to the Induction of Ferroptosis. JACS AU 2023; 3:1100-1117. [PMID: 37124288 PMCID: PMC10131203 DOI: 10.1021/jacsau.2c00681] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Revised: 02/14/2023] [Accepted: 02/14/2023] [Indexed: 05/03/2023]
Abstract
Ferroptosis is a form of regulated cell death driven by lipid peroxidation of polyunsaturated fatty acids (PUFAs). Lipid peroxidation can propagate through either the hydrogen-atom transfer (HAT) or peroxyl radical addition (PRA) mechanism. However, the contribution of the PRA mechanism to the induction of ferroptosis has not been studied. In this study, we aim to elucidate the relationship between the reactivity and mechanisms of lipid peroxidation and ferroptosis induction. We found that while some peroxidation-reactive lipids, such as 7-dehydrocholesterol, vitamins D3 and A, and coenzyme Q10, suppress ferroptosis, both nonconjugated and conjugated PUFAs enhanced cell death induced by RSL3, a ferroptosis inducer. Importantly, we found that conjugated PUFAs, including conjugated linolenic acid (CLA 18:3) and conjugated linoleic acid (CLA 18:2), can induce or potentiate ferroptosis much more potently than nonconjugated PUFAs. We next sought to elucidate the mechanism underlying the different ferroptosis-inducing potency of conjugated and nonconjugated PUFAs. Lipidomics revealed that conjugated and nonconjugated PUFAs are incorporated into distinct cellular lipid species. The different peroxidation mechanisms predict the formation of higher levels of reactive electrophilic aldehydes from conjugated PUFAs than nonconjugated PUFAs, which was confirmed by aldehyde-trapping and mass spectrometry. RNA sequencing revealed that protein processing in the endoplasmic reticulum and proteasome are among the most significantly upregulated pathways in cells treated with CLA 18:3, suggesting increased ER stress and activation of unfolded protein response. These results suggest that protein damage by lipid electrophiles is a key step in ferroptosis.
Collapse
Affiliation(s)
- Quynh Do
- Department
of Medicinal Chemistry, University of Washington, Seattle, Washington 98195, United States
| | - Rutan Zhang
- Department
of Medicinal Chemistry, University of Washington, Seattle, Washington 98195, United States
| | - Gavin Hooper
- Department
of Medicinal Chemistry, University of Washington, Seattle, Washington 98195, United States
| | - Libin Xu
- Department
of Medicinal Chemistry, University of Washington, Seattle, Washington 98195, United States
| |
Collapse
|
34
|
Singh A, Kukal S, Kanojia N, Singh M, Saso L, Kukreti S, Kukreti R. Lipid Mediated Brain Disorders: A Perspective. Prostaglandins Other Lipid Mediat 2023; 167:106737. [PMID: 37086954 DOI: 10.1016/j.prostaglandins.2023.106737] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Revised: 04/10/2023] [Accepted: 04/18/2023] [Indexed: 04/24/2023]
Abstract
The brain, one of the most resilient organs of the body is highly enriched in lipid content, suggesting the essential role of lipids in brain physiological activities. Lipids constitute an important structural part of the brain and act as a rich source of metabolic energy. Besides, lipids in their bioactive form (known as bioactive lipids) play an essential signaling and regulatory role, facilitating neurogenesis, synaptogenesis, and cell-cell communication. Brain lipid metabolism is thus a tightly regulated process. Any alteration/dysregulation of lipid metabolism greatly impact brain health and activity. Moreover, since central nervous system (CNS) is the most metabolically active system and lacks an efficient antioxidative defence system, it acts as a hub for the production of reactive oxygen species (ROS) and subsequent lipid peroxidation. These peroxidation events are reported during pathological changes such as neuronal tissue injury and inflammation. Present review is a modest attempt to gain insights into the role of dysregulated bioactive lipid levels and lipid oxidation status in the pathogenesis and progression of neurodegenerative disorders. This may open up new avenues exploiting lipids as the therapeutic targets for improving brain health, and treatment of nervous system disorders.
Collapse
Affiliation(s)
- Anju Singh
- Department of Chemistry, Ramjas College, University of Delhi, Delhi 110007, India; Nucleic Acids Research Lab, Department of Chemistry, University of Delhi (North Campus), Delhi 110007, India
| | - Samiksha Kukal
- Genomics and Molecular Medicine Unit, CSIR-Institute of Genomics and Integrative Biology (IGIB), Mall Road, Delhi 110007, India; Academy of Scientific and Innovative Research (AcSIR), CSIR-Institute of Genomics and Integrative Biology (CSIR-IGIB) Campus, Delhi 110007, India
| | - Neha Kanojia
- Genomics and Molecular Medicine Unit, CSIR-Institute of Genomics and Integrative Biology (IGIB), Mall Road, Delhi 110007, India; Academy of Scientific and Innovative Research (AcSIR), CSIR-Institute of Genomics and Integrative Biology (CSIR-IGIB) Campus, Delhi 110007, India
| | - Mahak Singh
- Department of Chemistry, Ramjas College, University of Delhi, Delhi 110007, India
| | - Luciano Saso
- Department of Physiology and Pharmacology "Vittorio Erspamer", Sapienza University of Rome, P. le Aldo Moro 5, 00185 Rome, Italy
| | - Shrikant Kukreti
- Nucleic Acids Research Lab, Department of Chemistry, University of Delhi (North Campus), Delhi 110007, India
| | - Ritushree Kukreti
- Genomics and Molecular Medicine Unit, CSIR-Institute of Genomics and Integrative Biology (IGIB), Mall Road, Delhi 110007, India; Academy of Scientific and Innovative Research (AcSIR), CSIR-Institute of Genomics and Integrative Biology (CSIR-IGIB) Campus, Delhi 110007, India.
| |
Collapse
|
35
|
Pandey SP, P K, Dutta T, Chakraborty B, Koner AL, Singh PK. Mitochondria-Directing Fluorogenic Probe: An Efficient Amyloid Marker for Imaging Lipid Metabolite-Induced Protein Aggregation in Live Cells and Caenorhabditis elegans. Anal Chem 2023; 95:6341-6350. [PMID: 37014217 DOI: 10.1021/acs.analchem.2c05466] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/05/2023]
Abstract
The design and development of optical probes for sensing neurotoxic amyloid fibrils are active and important areas of research and are undergoing continuous advancements. In this paper, we have synthesized a red emissive styryl chromone-based fluorophore (SC1) for fluorescence-based detection of amyloid fibrils. SC1 records exceptional modulation in its photophysical properties in the presence of amyloid fibrils, which has been attributed to the extreme sensitivity of its photophysical properties toward the immediate microenvironment of the probe in the fibrillar matrix. SC1 also shows very high selectivity toward the amyloid-aggregated form of the protein as compared to its native form. The probe is also able to monitor the kinetic progression of the fibrillation process, with comparable efficiency as that of the most popular amyloid probe, Thioflavin-T. Moreover, the performance of SC1 is least sensitive to the ionic strength of the medium, which is an advantage over Thioflavin-T. In addition, the molecular level interaction forces between the probe and the fibrillar matrix have been interrogated by molecular docking calculations which suggest the binding of the probe to the exterior channel of the fibrils. The probe has also been demonstrated to sense protein aggregates from the Aβ-40 protein, which is known to be responsible for Alzheimer's disease. Moreover, SC1 exhibited excellent biocompatibility and exclusive accumulation at mitochondria which allowed us to successfully demonstrate the applicability of this probe to detect mitochondrial-aggregated protein induced by an oxidative stress indicator molecule 4-hydroxy-2-nonenal (4-HNE) in A549 cell lines as well as in a simple animal model like Caenorhabditis elegans. Overall, the styryl chromone-based probe presents a potentially exciting alternative for the sensing of neurotoxic protein aggregation species both in vitro as well as in vivo.
Collapse
Affiliation(s)
- Shrishti P Pandey
- Department of Biotechnology, Mithibai College of Arts, Chauhan Institute of Science and Amrutben Jivanlal College of Commerce and Economics, Vile Parle (W) 400056, India
| | - Kavyashree P
- Bionanotechnology Laboratory, Department of Chemistry, Indian Institute of Science Education and Research Bhopal, Bhopal Bypass Road, Bhauri, Bhopal 462066, Madhya Pradesh, India
| | - Tanoy Dutta
- Bionanotechnology Laboratory, Department of Chemistry, Indian Institute of Science Education and Research Bhopal, Bhopal Bypass Road, Bhauri, Bhopal 462066, Madhya Pradesh, India
| | - Barsha Chakraborty
- Bionanotechnology Laboratory, Department of Chemistry, Indian Institute of Science Education and Research Bhopal, Bhopal Bypass Road, Bhauri, Bhopal 462066, Madhya Pradesh, India
| | - Apurba Lal Koner
- Bionanotechnology Laboratory, Department of Chemistry, Indian Institute of Science Education and Research Bhopal, Bhopal Bypass Road, Bhauri, Bhopal 462066, Madhya Pradesh, India
| | - Prabhat K Singh
- Radiation and Photochemistry Division, Bhabha Atomic Research Centre, Mumbai 400085, India
- Homi Bhabha National Institute, Anushaktinagar, Mumbai 400085, India
| |
Collapse
|
36
|
Milkovic L, Zarkovic N, Marusic Z, Zarkovic K, Jaganjac M. The 4-Hydroxynonenal–Protein Adducts and Their Biological Relevance: Are Some Proteins Preferred Targets? Antioxidants (Basel) 2023; 12:antiox12040856. [PMID: 37107229 PMCID: PMC10135105 DOI: 10.3390/antiox12040856] [Citation(s) in RCA: 19] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Revised: 03/27/2023] [Accepted: 03/29/2023] [Indexed: 04/05/2023] Open
Abstract
It is well known that oxidative stress and lipid peroxidation (LPO) play a role in physiology and pathology. The most studied LPO product with pleiotropic capabilities is 4-hydroxynonenal (4-HNE). It is considered as an important mediator of cellular signaling processes and a second messenger of reactive oxygen species. The effects of 4-HNE are mainly attributed to its adduction with proteins. Whereas the Michael adducts thus formed are preferred in an order of potency of cysteine > histidine > lysine over Schiff base formation, it is not known which proteins are the preferred targets for 4-HNE under what physiological or pathological conditions. In this review, we briefly discuss the methods used to identify 4-HNE–protein adducts, the progress of mass spectrometry in deciphering the specific protein targets, and their biological relevance, focusing on the role of 4-HNE protein adducts in the adaptive response through modulation of the NRF2/KEAP1 pathway and ferroptosis.
Collapse
Affiliation(s)
- Lidija Milkovic
- Laboratory for Oxidative Stress, Division of Molecular Medicine, Ruder Boskovic Institute, Bijenicka 54, 10000 Zagreb, Croatia
| | - Neven Zarkovic
- Laboratory for Oxidative Stress, Division of Molecular Medicine, Ruder Boskovic Institute, Bijenicka 54, 10000 Zagreb, Croatia
| | - Zlatko Marusic
- Division of Pathology, Clinical Hospital Centre Zagreb, Kispaticeva 12, 10000 Zagreb, Croatia
| | - Kamelija Zarkovic
- Division of Pathology, Clinical Hospital Centre Zagreb, Kispaticeva 12, 10000 Zagreb, Croatia
| | - Morana Jaganjac
- Laboratory for Oxidative Stress, Division of Molecular Medicine, Ruder Boskovic Institute, Bijenicka 54, 10000 Zagreb, Croatia
| |
Collapse
|
37
|
Jia Y, Hui L, Sun L, Guo D, Shi M, Zhang K, Yang P, Wang Y, Liu F, Shen O, Zhu Z. Association Between Human Blood Metabolome and the Risk of Psychiatric Disorders. Schizophr Bull 2023; 49:428-443. [PMID: 36124769 PMCID: PMC10016401 DOI: 10.1093/schbul/sbac130] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/14/2022]
Abstract
BACKGROUND AND HYPOTHESIS To identify promising drug targets for psychiatric disorders, we applied Mendelian randomization (MR) design to systematically screen blood metabolome for potential mediators of psychiatric disorders and further predict target-mediated side effects. STUDY DESIGN We selected 92 unique blood metabolites from 3 metabolome genome-wide association studies (GWASs) with totally 147 827 participants. Summary statistics for bipolar disorder (BIP), attention deficit hyperactivity disorder (ADHD), obsessive-compulsive disorder (OCD), major depressive disorder (MDD), schizophrenia (SCZ), panic disorder (PD), autistic spectrum disorder (ASD), and anorexia nervosa (AN) originated from the Psychiatric Genomics Consortium, involving 1 143 340 participants. Mendelian randomization (MR) analyses were conducted to estimate associations of blood metabolites with psychiatric disorders. Phenome-wide MR analysis was further performed to predict side effects mediated by metabolite-targeted interventions. RESULTS Eight metabolites were identified associated with psychiatric disorders, including five established mediators: N-acetylornithine (BIP: OR, 0.72 [95% CI, 0.66-0.79]; SCZ: OR, 0.74 [0.64-0.84]), glycine (BIP: OR, 0.62 [0.50-0.77]), docosahexaenoic acid (MDD: OR, 0.96 [0.94-0.97]), 3-Hydroxybutyrate (MDD: OR, 1.14 [1.08-1.21]), butyrylcarnitine (SCZ: OR, 1.22 [1.12-1.32]); and three novel mediators: 1-arachidonoylglycerophosphocholine (1-arachidonoyl-GPC)(BIP: OR, 0.31 [0.23-0.41]), glycoproteins (BIP: OR, 0.94 [0.92-0.97]), sphingomyelins (AN: OR, 1.12 [1.06-1.19]). Phenome-wide MR analysis showed that all identified metabolites except for N-acetylornithine and 3-Hydroxybutyrate had additional effects on nonpsychiatric diseases, while glycine, 3-Hydroxybutyrate, N-acetylornithine, and butyrylcarnitine had no adverse side effects. CONCLUSIONS This MR study identified five established and three novel mediators for psychiatric disorders. N-acetylornithine, glycine, 3-Hydroxybutyrate, and butyrylcarnitine might be promising targets against psychiatric disorders with no predicted adverse side effects.
Collapse
Affiliation(s)
- Yiming Jia
- Department of Epidemiology, School of Public Health and Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Medical College of Soochow University, Suzhou, China
| | - Li Hui
- Research Center of Biological Psychiatry, The Affiliated Guangji Hospital of Soochow University, Suzhou, China
| | - Lulu Sun
- Department of Epidemiology, School of Public Health and Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Medical College of Soochow University, Suzhou, China
| | - Daoxia Guo
- Department of Epidemiology, School of Public Health and Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Medical College of Soochow University, Suzhou, China
- School of Nursing, Medical College of Soochow University, Suzhou, China
| | - Mengyao Shi
- Department of Epidemiology, School of Public Health and Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Medical College of Soochow University, Suzhou, China
| | - Kaixin Zhang
- Department of Epidemiology, School of Public Health and Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Medical College of Soochow University, Suzhou, China
| | - Pinni Yang
- Department of Epidemiology, School of Public Health and Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Medical College of Soochow University, Suzhou, China
| | - Yu Wang
- Department of Epidemiology, School of Public Health and Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Medical College of Soochow University, Suzhou, China
| | - Fanghua Liu
- Department of Epidemiology, School of Public Health and Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Medical College of Soochow University, Suzhou, China
| | - Ouxi Shen
- Department of Occupational Health, Suzhou Industrial Park Center for Disease Control and Prevention, Suzhou, China
| | - Zhengbao Zhu
- Department of Epidemiology, School of Public Health and Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Medical College of Soochow University, Suzhou, China
| |
Collapse
|
38
|
Liu X, Bai M, Fan L, Lou Z. Serum 4-hydroxynonenal associates with the recurrence of patients with primary cerebral infarction. Front Cell Neurosci 2022; 16:998512. [DOI: 10.3389/fncel.2022.998512] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Accepted: 10/18/2022] [Indexed: 11/11/2022] Open
Abstract
Background4-Hydroxynonenal (4-HNE), an α, β-unsaturated hydroxyalkenal, has been found to be associated with aspirin resistance, which is a risk factor for recurrent cerebral infarction. However, its effect on recurrent cerebral infarction is less defined. We designed this study to investigate the association between 4-HNE and increased risk of recurrent cerebral infarction.MethodsWe recruited 189 patients with primary cerebral infarction from 2017 to 2019. According to the recurrence of cerebral infarction during the 3-year follow-up period, they were divided into two groups, namely, the non-recurrence group (n = 93) and the recurrence group (n = 96). All patients were analyzed to explore the risk factors for the recurrence of primary cerebral infarction and the predictive value of serum 4-HNE for the recurrence of cerebral infarction.ResultsThe levels of serum 4-HNE in patients of the recurrence group were significantly higher than that in patients of the non-recurrence group. There was a positive correlation between serum 4-HNE levels and the serum levels of triglyceride (r = 0.448, p = 0.008) and low-density lipoprotein cholesterol (LDL-C; r = 0.442, p = 0.002) in primary cerebral infarction patients. Cox proportional hazards modeling showed that demographic and certain clinical parameters, such as age, serum triglyceride levels, the National Institutes of Health Stroke Scale (NIHSS) scores, and serum 4-HNE levels, were independent factors for the recurrence in patients. The results of the receiver operating characteristic (ROC) curve showed that the area under the curve (AUC) value of serum 4-HNE in patients with cerebral infarction recurrence was 0.703, and when the cutoff value of serum 4-HNE was set at 42.34 ng/ml, the sensitivity and specificity values of serum 4-HNE in predicting recurrent cerebral infarction were 79.20 and 52.70%, respectively.ConclusionSerum 4-HNE is an independent risk factor for the recurrence of patients with primary cerebral infarction, and it may become a new intervention way to prevent the recurrence of patients with cerebral infarction.
Collapse
|
39
|
Stress and viral insults do not trigger E200K PrP conversion in human cerebral organoids. PLoS One 2022; 17:e0277051. [PMID: 36301953 PMCID: PMC9612459 DOI: 10.1371/journal.pone.0277051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Accepted: 10/18/2022] [Indexed: 12/03/2022] Open
Abstract
Prion diseases are a group of rare, transmissible, and invariably fatal neurodegenerative diseases that affect both humans and animals. The cause of these diseases is misfolding of the prion protein into pathological isoforms called prions. Of all human prion diseases, 10-15% of cases are genetic and the E200K mutation, which causes familial Creutzfeldt-Jakob disease (CJD), is the most prevalent. For both sporadic and genetic disease, it remains uncertain as to how initial protein misfolding is triggered. Prior studies have linked protein misfolding with oxidative stress insults, deregulated interactions with cellular cofactors, and viral infections. Our previous work developed a cerebral organoid (CO) model using human induced pluripotent stem cells containing the E200K mutation. COs are three-dimensional human neural tissues that permit the study of host genetics and environmental factors that contribute to disease onset. Isogenically matched COs with and without the E200K mutation were used to investigate the propensity of E200K PrP to misfold following cellular insults associated with oxidative stress. Since viral infections have also been associated with oxidative stress and neurodegenerative diseases, we additionally investigated the influence of Herpes Simplex Type-1 virus (HSV1), a neurotropic virus that establishes life-long latent infection in its host, on E200K PrP misfolding. While COs proved to be highly infectable with HSV1, neither acute nor latent infection, or direct oxidative stress insult, resulted in evidence of E200K prion misfolding. We conclude that misfolding into seeding-active PrP species is not readily induced by oxidative stress or HSV1 in our organoid system.
Collapse
|
40
|
Electrophilic Aldehyde 4-Hydroxy-2-Nonenal Mediated Signaling and Mitochondrial Dysfunction. Biomolecules 2022; 12:biom12111555. [PMID: 36358905 PMCID: PMC9687674 DOI: 10.3390/biom12111555] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Revised: 10/22/2022] [Accepted: 10/23/2022] [Indexed: 01/21/2023] Open
Abstract
Reactive oxygen species (ROS), a by-product of aerobic life, are highly reactive molecules with unpaired electrons. The excess of ROS leads to oxidative stress, instigating the peroxidation of polyunsaturated fatty acids (PUFA) in the lipid membrane through a free radical chain reaction and the formation of the most bioactive aldehyde, known as 4-hydroxynonenal (4-HNE). 4-HNE functions as a signaling molecule and toxic product and acts mainly by forming covalent adducts with nucleophilic functional groups in proteins, nucleic acids, and lipids. The mitochondria have been implicated as a site for 4-HNE generation and adduction. Several studies clarified how 4-HNE affects the mitochondria's functions, including bioenergetics, calcium homeostasis, and mitochondrial dynamics. Our research group has shown that 4-HNE activates mitochondria apoptosis-inducing factor (AIFM2) translocation and facilitates apoptosis in mice and human heart tissue during anti-cancer treatment. Recently, we demonstrated that a deficiency of SOD2 in the conditional-specific cardiac knockout mouse increases ROS, and subsequent production of 4-HNE inside mitochondria leads to the adduction of several mitochondrial respiratory chain complex proteins. Moreover, we highlighted the physiological functions of HNE and discussed their relevance in human pathophysiology and current discoveries concerning 4-HNE effects on mitochondria.
Collapse
|
41
|
Žarković N, Orehovec B, Baršić B, Tarle M, Kmet M, Lukšić I, Tatzber F, Wonisch W, Skrzydlewska E, Łuczaj W. Lipidomics Revealed Plasma Phospholipid Profile Differences between Deceased and Recovered COVID-19 Patients. Biomolecules 2022; 12:biom12101488. [PMID: 36291697 PMCID: PMC9599609 DOI: 10.3390/biom12101488] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Revised: 10/06/2022] [Accepted: 10/13/2022] [Indexed: 01/08/2023] Open
Abstract
Thorough understanding of metabolic changes, including lipidome alteration, associated with the development of COVID-19 appears to be crucial, as new types of coronaviruses are still reported. In this study, we analyzed the differences in the plasma phospholipid profiles of the deceased COVID-19 patients, those who recovered and healthy people. Due to identified abnormalities in plasma phospholipid profiles, deceased patients were further divided into two subgroups (D1 and D2). Increased levels of phosphatidylethanolamines (PE), phosphatidylcholines (PC) and phosphatidylserines (PS) were found in the plasma of recovered patients and the majority of deceased patients (first subgroup D1) compared to the control group. However, abundances of all relevant PE, PC and PS species decreased dramatically in the plasma of the second subgroup (D2) of five deceased patients. These patients also had significantly decreased plasma COX-2 activity when compared to the control, in contrast to unchanged and increased COX-2 activity in the plasma of the other deceased patients and recovered patients, respectively. Moreover, these five deceased patients were characterized by abnormally low CRP levels and tremendous increase in LDH levels, which may be the result of other pathophysiological disorders, including disorders of the immune system, liver damage and haemolytic anemia. In addition, an observed trend to decrease the autoantibodies against oxidative modifications of low-density lipoprotein (oLAb) titer in all, especially in deceased patients, indicate systemic oxidative stress and altered immune system that may have prognostic value in COVID-19.
Collapse
Affiliation(s)
- Neven Žarković
- Ruđer Bošković Institute, Laboratory for Oxidative Stress, 10000 Zagreb, Croatia
- Correspondence:
| | | | - Bruno Baršić
- Clinical Hospital Dubrava, 10000 Zagreb, Croatia
| | - Marko Tarle
- Clinical Hospital Dubrava, 10000 Zagreb, Croatia
| | - Marta Kmet
- Clinical Hospital Dubrava, 10000 Zagreb, Croatia
| | - Ivica Lukšić
- Clinical Hospital Dubrava, 10000 Zagreb, Croatia
- Department of Pathology, University of Zagreb School of Medicine, 10000 Zagreb, Croatia
| | - Franz Tatzber
- Omnignostica Ltd., 3421 Höflein an der Donau, Austria
| | | | - Elżbieta Skrzydlewska
- Department of Analytical Chemistry, Medical University of Bialystok, A. Mickiewicza 2D, 15-222 Bialystok, Poland
| | - Wojciech Łuczaj
- Department of Analytical Chemistry, Medical University of Bialystok, A. Mickiewicza 2D, 15-222 Bialystok, Poland
| |
Collapse
|
42
|
Changing Perspectives from Oxidative Stress to Redox Signaling-Extracellular Redox Control in Translational Medicine. Antioxidants (Basel) 2022; 11:antiox11061181. [PMID: 35740078 PMCID: PMC9228063 DOI: 10.3390/antiox11061181] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Revised: 06/08/2022] [Accepted: 06/10/2022] [Indexed: 12/07/2022] Open
Abstract
Extensive research has changed the understanding of oxidative stress that has been linked to every major disease. Today we distinguish oxidative eu- and distress, acknowledging that redox modifications are crucial for signal transduction in the form of specific thiol switches. Long underestimated, reactive species and redox proteins of the Thioredoxin (Trx) family are indeed essential for physiological processes. Moreover, extracellular redox proteins, low molecular weight thiols and thiol switches affect signal transduction and cell–cell communication. Here, we highlight the impact of extracellular redox regulation for health, intermediate pathophenotypes and disease. Of note, recent advances allow the analysis of redox changes in body fluids without using invasive and expensive techniques. With this new knowledge in redox biochemistry, translational strategies can lead to innovative new preventive and diagnostic tools and treatments in life sciences and medicine.
Collapse
|
43
|
Rwere F, Yu X, Chen CH, Gross ER. Aldehydes, Aldehyde Metabolism, and the ALDH2 Consortium. Biomolecules 2022; 12:763. [PMID: 35740888 PMCID: PMC9221099 DOI: 10.3390/biom12060763] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Accepted: 05/26/2022] [Indexed: 12/20/2022] Open
Abstract
The discovery of aldehydes dates back to 1774 when Carl Wilhelm Scheele synthesized acetaldehyde [...].
Collapse
Affiliation(s)
- Freeborn Rwere
- Department of Anesthesiology, Perioperative and Pain Medicine, School of Medicine, Stanford University, Stanford, CA 94305, USA; (F.R.); (X.Y.)
| | - Xuan Yu
- Department of Anesthesiology, Perioperative and Pain Medicine, School of Medicine, Stanford University, Stanford, CA 94305, USA; (F.R.); (X.Y.)
| | - Che-Hong Chen
- Department of Chemical and Systems Biology, School of Medicine, Stanford University, Stanford, CA 94305, USA
| | - Eric R. Gross
- Department of Anesthesiology, Perioperative and Pain Medicine, School of Medicine, Stanford University, Stanford, CA 94305, USA; (F.R.); (X.Y.)
| |
Collapse
|
44
|
Ferreri C, Sansone A, Chatgilialoglu C, Ferreri R, Amézaga J, Burgos MC, Arranz S, Tueros I. Critical Review on Fatty Acid-Based Food and Nutraceuticals as Supporting Therapy in Cancer. Int J Mol Sci 2022; 23:ijms23116030. [PMID: 35682708 PMCID: PMC9181022 DOI: 10.3390/ijms23116030] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Revised: 05/17/2022] [Accepted: 05/24/2022] [Indexed: 02/02/2023] Open
Abstract
Fatty acids have an important place in both biological and nutritional contexts and, from a clinical point of view, they have known consequences for diseases’ onset and development, including cancer. The use of fatty acid-based food and nutraceuticals to support cancer therapy is a multidisciplinary subject, involving molecular and clinical research. Knowledge regarding polyunsaturated fatty acids essentiality/oxidizability and the role of lipogenesis-desaturase pathways for cell growth, as well as oxidative reactivity in cancer cells, are discussed, since they can drive the choice of fatty acids using their multiple roles to support antitumoral drug activity. The central role of membrane fatty acid composition is highlighted for the application of membrane lipid therapy. As fatty acids are also known as biomarkers of cancer onset and progression, the personalization of the fatty acid-based therapy is also possible, taking into account other important factors such as formulation, bioavailability and the distribution of the supplementation. A holistic approach emerges combining nutra- and pharma-strategies in an appropriate manner, to develop further knowledge and applications in cancer therapy.
Collapse
Affiliation(s)
- Carla Ferreri
- Istituto per la Sintesi Organica e la Fotoreattività, Consiglio Nazionale delle Ricerche, Via Piero Gobetti 101, 40129 Bologna, Italy; (A.S.); (C.C.)
- Correspondence:
| | - Anna Sansone
- Istituto per la Sintesi Organica e la Fotoreattività, Consiglio Nazionale delle Ricerche, Via Piero Gobetti 101, 40129 Bologna, Italy; (A.S.); (C.C.)
| | - Chryssostomos Chatgilialoglu
- Istituto per la Sintesi Organica e la Fotoreattività, Consiglio Nazionale delle Ricerche, Via Piero Gobetti 101, 40129 Bologna, Italy; (A.S.); (C.C.)
| | - Rosaria Ferreri
- Department of Integrated Medicine, Tuscany Reference Centre for Integrated Medicine in the Hospital Pathway, Pitigliano Hospital, ASL Sudest Toscana, 58017 Pitigliano, Italy;
| | - Javier Amézaga
- AZTI, Food Research, Basque Research and Technology Alliance (BRTA), Parque Tecnológico de Bizkaia, Astondo Bidea, Edificio 609, 48160 Derio, Spain; (J.A.); (M.C.B.); (S.A.); (I.T.)
| | - Mercedes Caro Burgos
- AZTI, Food Research, Basque Research and Technology Alliance (BRTA), Parque Tecnológico de Bizkaia, Astondo Bidea, Edificio 609, 48160 Derio, Spain; (J.A.); (M.C.B.); (S.A.); (I.T.)
| | - Sara Arranz
- AZTI, Food Research, Basque Research and Technology Alliance (BRTA), Parque Tecnológico de Bizkaia, Astondo Bidea, Edificio 609, 48160 Derio, Spain; (J.A.); (M.C.B.); (S.A.); (I.T.)
| | - Itziar Tueros
- AZTI, Food Research, Basque Research and Technology Alliance (BRTA), Parque Tecnológico de Bizkaia, Astondo Bidea, Edificio 609, 48160 Derio, Spain; (J.A.); (M.C.B.); (S.A.); (I.T.)
| |
Collapse
|
45
|
Sharma S, Patel F, Ara H, Bess E, Shum A, Bhattarai S, Subedi U, Bell DS, Bhuiyan MS, Sun H, Batinic-Haberle I, Panchatcharam M, Miriyala S. Rotenone-Induced 4-HNE Aggresome Formation and Degradation in HL-1 Cardiomyocytes: Role of Autophagy Flux. Int J Mol Sci 2022; 23:ijms23094675. [PMID: 35563066 PMCID: PMC9105393 DOI: 10.3390/ijms23094675] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Revised: 04/21/2022] [Accepted: 04/22/2022] [Indexed: 11/22/2022] Open
Abstract
Reactive oxygen species (ROS) cause oxidative stress by generating reactive aldehydes known as 4-hydroxynonenal (4-HNE). 4-HNE modifies protein via covalent adduction; however, little is known about the degradation mechanism of 4-HNE-adducted proteins. Autophagy is a dynamic process that maintains cellular homeostasis by removing damaged organelles and proteins. In this study, we determined the role of a superoxide dismutase (SOD) mimetic MnTnBuOE-2-PyP5+ (MnP, BMX-001) on rotenone-induced 4-HNE aggresome degradation in HL-1 cardiomyocytes. A rotenone treatment (500 nM) given for 24 h demonstrated both increased ROS and 4-HNE aggresome accumulation in HL-1 cardiomyocytes. In addition, cardiomyocytes treated with rotenone displayed an increase in the autophagy marker LC3-II, as shown by immunoblotting and immunofluorescence. A pre-treatment with MnP (20 µM) for 24 h attenuated rotenone-induced ROS formation. An MnP pre-treatment showed decreased 4-HNE aggresomes and LC3-II formation. A rotenone-induced increase in autophagosomes was attenuated by a pre-treatment with MnP, as shown by fluorescent-tagged LC3 (tfLC3). Rotenone increased tubulin hyperacetylation through the ROS-mediated pathway, which was attenuated by MnP. The disruption of autophagy caused HL-1 cell death because a 3-methyladenine inhibitor of autophagosomes caused reduced cell death. Yet, rapamycin, an inducer of autophagy, increased cell death. These results indicated that a pre-treatment with MnP decreased rotenone-induced 4-HNE aggresomes by enhancing the degradation process.
Collapse
Affiliation(s)
- Sudha Sharma
- Department of Cellular Biology and Anatomy, Louisiana State University Health Sciences Center, Shreveport, LA 71103, USA; (S.S.); (F.P.); (H.A.); (E.B.); (A.S.); (S.B.); (U.S.); (D.S.B.); (H.S.)
| | - Foram Patel
- Department of Cellular Biology and Anatomy, Louisiana State University Health Sciences Center, Shreveport, LA 71103, USA; (S.S.); (F.P.); (H.A.); (E.B.); (A.S.); (S.B.); (U.S.); (D.S.B.); (H.S.)
| | - Hosne Ara
- Department of Cellular Biology and Anatomy, Louisiana State University Health Sciences Center, Shreveport, LA 71103, USA; (S.S.); (F.P.); (H.A.); (E.B.); (A.S.); (S.B.); (U.S.); (D.S.B.); (H.S.)
| | - Ezra Bess
- Department of Cellular Biology and Anatomy, Louisiana State University Health Sciences Center, Shreveport, LA 71103, USA; (S.S.); (F.P.); (H.A.); (E.B.); (A.S.); (S.B.); (U.S.); (D.S.B.); (H.S.)
| | - Alika Shum
- Department of Cellular Biology and Anatomy, Louisiana State University Health Sciences Center, Shreveport, LA 71103, USA; (S.S.); (F.P.); (H.A.); (E.B.); (A.S.); (S.B.); (U.S.); (D.S.B.); (H.S.)
| | - Susmita Bhattarai
- Department of Cellular Biology and Anatomy, Louisiana State University Health Sciences Center, Shreveport, LA 71103, USA; (S.S.); (F.P.); (H.A.); (E.B.); (A.S.); (S.B.); (U.S.); (D.S.B.); (H.S.)
| | - Utsab Subedi
- Department of Cellular Biology and Anatomy, Louisiana State University Health Sciences Center, Shreveport, LA 71103, USA; (S.S.); (F.P.); (H.A.); (E.B.); (A.S.); (S.B.); (U.S.); (D.S.B.); (H.S.)
| | - Daquonte Sanard Bell
- Department of Cellular Biology and Anatomy, Louisiana State University Health Sciences Center, Shreveport, LA 71103, USA; (S.S.); (F.P.); (H.A.); (E.B.); (A.S.); (S.B.); (U.S.); (D.S.B.); (H.S.)
| | - Md. Shenuarin Bhuiyan
- Department of Pathology and Translational Pathobiology, Louisiana State University Health Sciences Center, Shreveport, LA 71103, USA;
| | - Hong Sun
- Department of Cellular Biology and Anatomy, Louisiana State University Health Sciences Center, Shreveport, LA 71103, USA; (S.S.); (F.P.); (H.A.); (E.B.); (A.S.); (S.B.); (U.S.); (D.S.B.); (H.S.)
| | - Ines Batinic-Haberle
- Department of Radiation Oncology, Duke University School of Medicine, Durham, NC 27710, USA;
| | - Manikandan Panchatcharam
- Department of Cellular Biology and Anatomy, Louisiana State University Health Sciences Center, Shreveport, LA 71103, USA; (S.S.); (F.P.); (H.A.); (E.B.); (A.S.); (S.B.); (U.S.); (D.S.B.); (H.S.)
- Correspondence: (M.P.); (S.M.); Tel.: +1-3-186-756-938 (M.P.); +1-3-186-758-326 (S.M.)
| | - Sumitra Miriyala
- Department of Cellular Biology and Anatomy, Louisiana State University Health Sciences Center, Shreveport, LA 71103, USA; (S.S.); (F.P.); (H.A.); (E.B.); (A.S.); (S.B.); (U.S.); (D.S.B.); (H.S.)
- Correspondence: (M.P.); (S.M.); Tel.: +1-3-186-756-938 (M.P.); +1-3-186-758-326 (S.M.)
| |
Collapse
|
46
|
Zhu W, Feng D, Shi X, Wei Q, Yang L. The Potential Role of Mitochondrial Acetaldehyde Dehydrogenase 2 in Urological Cancers From the Perspective of Ferroptosis and Cellular Senescence. Front Cell Dev Biol 2022; 10:850145. [PMID: 35517510 PMCID: PMC9065557 DOI: 10.3389/fcell.2022.850145] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Accepted: 03/21/2022] [Indexed: 12/21/2022] Open
Abstract
Overproduction of reactive oxygen species (ROS) and superlative lipid peroxidation promote tumorigenesis, and mitochondrial aldehyde dehydrogenase 2 (ALDH2) is associated with the detoxification of ROS-mediated lipid peroxidation-generated reactive aldehydes such as 4-hydroxy-2-nonenal (4-HNE), malondialdehyde, and acrolein due to tobacco smoking. ALDH2 has been demonstrated to be highly associated with the prognosis and chemoradiotherapy sensitivity of many types of cancer, including leukemia, lung cancer, head and neck cancer, esophageal cancer, hepatocellular cancer, pancreatic cancer, and ovarian cancer. In this study, we explored the possible relationship between ALDH2 and urological cancers from the aspects of ferroptosis, epigenetic alterations, proteostasis, mitochondrial dysfunction, and cellular senescence.
Collapse
Affiliation(s)
| | | | | | - Qiang Wei
- *Correspondence: Qiang Wei, ; Lu Yang,
| | - Lu Yang
- *Correspondence: Qiang Wei, ; Lu Yang,
| |
Collapse
|
47
|
Malakul W, Seenak P, Jumroon N, Arikit S, Kumphune S, Nernpermpisooth N. Novel Coconut Vinegar Attenuates Hepatic and Vascular Oxidative Stress in Rats Fed a High-Cholesterol Diet. Front Nutr 2022; 9:835278. [PMID: 35356733 PMCID: PMC8959456 DOI: 10.3389/fnut.2022.835278] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Accepted: 01/24/2022] [Indexed: 12/20/2022] Open
Abstract
Background Hypercholesterolemia is an independent modifiable risk factor that accelerates the development of both non-alcoholic fatty liver and atherosclerosis. Coconut water contains a variety of phytochemicals that make it appealing for producing vinegar. Coconut vinegar is rapidly gaining popularity for health benefits in Southeast Asia. The purpose of this study is to evaluate the effect of daily supplementation of coconut vinegar on hepatic and vascular oxidative stress in rats fed a high-cholesterol diet (HCD). Methods Mature coconut water was fermented with coconut sap sugar using Saccharomyces cerevisiae and Acetobacter aceti vat Europeans, respectively. Bioactive compounds and antioxidant capacity of coconut vinegar were examined in vitro. Adult male Sprague-Dawley rats were randomly divided into four groups; the control group fed a standard diet (S), a group that received HCD (SC), a group that received HCD supplemented with coconut vinegar at a dose of 1 mL/kg/day (SCV), and a group that received HCD with atorvastatin at a dose of 30 mg/kg/day (SCA). After 8 weeks, serum metabolic profiles, fatty liver, hepatic, and vascular oxidative stress were determined. Results In in vitro studies, coconut vinegar was rich in phenolic compounds and organic acids. The antioxidant capacity of 30 μL of coconut vinegar was 181.55 ± 8.15 μM Trolox equivalent antioxidant capacity (TEAC). In the HCD fed rats, daily supplementation of coconut vinegar reduced weight gain, serum triglycerides, and fasting blood sugar levels without renal or liver toxicity. In the liver, coconut vinegar reduced the accumulation of both hepatic cholesterol and hepatic triglyceride, and it also reduced hepatic 4-hydroxynonenal (4-HNE) lipid peroxidation. In the aortic tissues, coconut vinegar increased nitric oxide bioavailability and reduced aortic 4-HNE lipid peroxidation. Conclusion Novel coconut vinegar is the source of antioxidants, and daily supplementation of coconut vinegar was found to attenuate dyslipidemia-induced hepatic and vascular oxidative stress by protective against cellular lipid peroxidation.
Collapse
Affiliation(s)
- Wachirawadee Malakul
- Department of Physiology, Faculty of Medical Sciences, Naresuan University, Phitsanulok, Thailand
| | - Porrnthanate Seenak
- Integrative Biomedical Research Unit (IBRU), Faculty of Allied Health Sciences, Naresuan University, Phitsanulok, Thailand
- Department of Cardio-Thoracic Technology, Faculty of Allied Health Sciences, Naresuan University, Phitsanulok, Thailand
| | - Noppadon Jumroon
- Integrative Biomedical Research Unit (IBRU), Faculty of Allied Health Sciences, Naresuan University, Phitsanulok, Thailand
- Department of Medical Technology, Faculty of Allied Health Sciences, Naresuan University, Phitsanulok, Thailand
| | - Siwaret Arikit
- Department of Agronomy, Faculty of Agriculture at Kamphaeng Saen, Kasetsart University, Kamphaeng Saen Campus, Nakhon Pathom, Thailand
| | - Sarawut Kumphune
- Integrative Biomedical Research Unit (IBRU), Faculty of Allied Health Sciences, Naresuan University, Phitsanulok, Thailand
- Biomedical Engineering Institute (BMEI), Chiang Mai University, Chiang Mai, Thailand
| | - Nitirut Nernpermpisooth
- Integrative Biomedical Research Unit (IBRU), Faculty of Allied Health Sciences, Naresuan University, Phitsanulok, Thailand
- Department of Cardio-Thoracic Technology, Faculty of Allied Health Sciences, Naresuan University, Phitsanulok, Thailand
| |
Collapse
|
48
|
Zanoni M, Bravaccini S, Fabbri F, Arienti C. Emerging Roles of Aldehyde Dehydrogenase Isoforms in Anti-cancer Therapy Resistance. Front Med (Lausanne) 2022; 9:795762. [PMID: 35299840 PMCID: PMC8920988 DOI: 10.3389/fmed.2022.795762] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Accepted: 02/10/2022] [Indexed: 12/19/2022] Open
Abstract
Aldehyde dehydrogenases (ALDHs) are a family of detoxifying enzymes often upregulated in cancer cells and associated with therapeutic resistance. In humans, the ALDH family comprises 19 isoenzymes active in the majority of mammalian tissues. Each ALDH isoform has a specific differential expression pattern and most of them have individual functional roles in cancer. ALDHs are overexpressed in subpopulations of cancer cells with stem-like features, where they are involved in several processes including cellular proliferation, differentiation, detoxification and survival, participating in lipids and amino acid metabolism and retinoic acid synthesis. In particular, ALDH enzymes protect cancer cells by metabolizing toxic aldehydes in less reactive and more soluble carboxylic acids. High metabolic activity as well as conventional anticancer therapies contribute to aldehyde accumulation, leading to DNA double strand breaks (DSB) through the generation of reactive oxygen species (ROS) and lipid peroxidation. ALDH overexpression is crucial not only for the survival of cancer stem cells but can also affect immune cells of the tumour microenvironment (TME). The reduction of ROS amount and the increase in retinoic acid signaling impairs immunogenic cell death (ICD) inducing the activation and stability of immunosuppressive regulatory T cells (Tregs). Dissecting the role of ALDH specific isoforms in the TME can open new scenarios in the cancer treatment. In this review, we summarize the current knowledge about the role of ALDH isoforms in solid tumors, in particular in association with therapy-resistance.
Collapse
Affiliation(s)
- Michele Zanoni
- Biosciences Laboratory,IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, Meldola, Italy
| | | | | | - Chiara Arienti
- Biosciences Laboratory,IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, Meldola, Italy
| |
Collapse
|
49
|
Kozłowska B, Sochanowicz B, Kraj L, Palusińska M, Kołsut P, Szymański Ł, Lewicki S, Śmigielski W, Kruszewski M, Leszek P. Expression of Iron Metabolism Proteins in Patients with Chronic Heart Failure. J Clin Med 2022; 11:jcm11030837. [PMID: 35160288 PMCID: PMC8837054 DOI: 10.3390/jcm11030837] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Revised: 01/28/2022] [Accepted: 02/03/2022] [Indexed: 11/25/2022] Open
Abstract
In heart failure, iron deficiency is a common comorbid disease that negatively influences exercise tolerance, number of hospitalizations and mortality rate, and this is why iron iv supplementation is recommended. Little is known about the changes in iron-related proteins in the human HF myocardium. The purpose of this study was to assess iron-related proteins in non-failing (NFH) vs. failing (FH) human myocardium. The study group consisted of 58 explanted FHs; control consisted of 31 NFHs unsuitable for transplantation. Myocardial proteins expressions: divalent metal transporter (DMT-1); L-type calcium channel (L-CH); transferrin receptors (TfR-1/TfR-2); ferritins: heavy (FT-H) or light (FT-L) chain, mitochondrial (FT-MT); ferroportin (FPN), regulatory factors and oxidative stress marker: 4-hydroxynonenal (4-HNE). In FH, the expression in almost all proteins responsible for iron transport: DMT-1, TfR-1, L-CH, except TfR-2, and storage: FT-H/-L/-MT were reduced, with no changes in FPN. Moreover, 4-HNE expression (pg/mg; NFH 10.6 ± 8.4 vs. FH 55.7 ± 33.7; p < 0.0001) in FH was increased. HNE-4 significantly correlated with DMT-1 (r = −0.377, p = 0.036), L-CH (r = −0.571, p = 0.001), FT-H (r = −0.379, p = 0.036), also FPN (r = 0.422, p = 0.018). Reducing iron-gathering proteins and elevated oxidative stress in failing hearts is very unfavorable for myocardiocytes. It should be taken into consideration before treatment with drugs or supplements that elevate free oxygen radicals in the heart.
Collapse
Affiliation(s)
- Bogna Kozłowska
- Department of Heart Failure and Transplantology, The Cardinal Stefan Wyszyński National Institute of Cardiology, Alpejska 42, 04-628 Warsaw, Poland;
| | - Barbara Sochanowicz
- Centre of Radiobiology and Biological Dosimetry, Institute of Nuclear Chemistry and Technology, Dorodna 16, 03-195 Warszawa, Poland; (B.S.); (M.K.)
| | - Leszek Kraj
- Department of Oncology, Medical University of Warsaw, 01-163 Warsaw, Poland;
- Department of Molecular Biology, Institute of Genetics and Animal Biotechnology, Polish Academy of Science, Postępu 36A, 05-552 Magdalenka, Poland; (M.P.); (Ł.S.); (S.L.)
| | - Małgorzata Palusińska
- Department of Molecular Biology, Institute of Genetics and Animal Biotechnology, Polish Academy of Science, Postępu 36A, 05-552 Magdalenka, Poland; (M.P.); (Ł.S.); (S.L.)
| | - Piotr Kołsut
- Department of Cardiac Surgery and Transplantology, The Cardinal Stefan Wyszyński National Institute of Cardiology, Alpejska 42, 04-628 Warsaw, Poland;
| | - Łukasz Szymański
- Department of Molecular Biology, Institute of Genetics and Animal Biotechnology, Polish Academy of Science, Postępu 36A, 05-552 Magdalenka, Poland; (M.P.); (Ł.S.); (S.L.)
| | - Sławomir Lewicki
- Department of Molecular Biology, Institute of Genetics and Animal Biotechnology, Polish Academy of Science, Postępu 36A, 05-552 Magdalenka, Poland; (M.P.); (Ł.S.); (S.L.)
- Faculty of Medical Sciences and Health Sciences, Kazimierz Pulaski University of Technology and Humanities, 26-600 Radom, Poland
| | - Witold Śmigielski
- Department of Epidemiology, Cardiovascular Disease Prevention and Health Promotion, The Cardinal Stefan Wyszyński National Institute of Cardiology, Alpejska 42, 04-628 Warsaw, Poland;
| | - Marcin Kruszewski
- Centre of Radiobiology and Biological Dosimetry, Institute of Nuclear Chemistry and Technology, Dorodna 16, 03-195 Warszawa, Poland; (B.S.); (M.K.)
- Department of Molecular Biology and Translational Research, Institute of Rural Health, Jaczewskiego 2, 20-090 Lublin, Poland
| | - Przemysław Leszek
- Department of Heart Failure and Transplantology, The Cardinal Stefan Wyszyński National Institute of Cardiology, Alpejska 42, 04-628 Warsaw, Poland;
- Correspondence: ; Tel.: +48-22-3434-483
| |
Collapse
|
50
|
Badiei A, Beltran WA, Aguirre GD. Altered transsulfuration pathway enzymes and redox homeostasis in inherited retinal degenerative diseases. Exp Eye Res 2022; 215:108902. [PMID: 34954206 PMCID: PMC8923955 DOI: 10.1016/j.exer.2021.108902] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Revised: 12/04/2021] [Accepted: 12/15/2021] [Indexed: 02/03/2023]
Abstract
Retinal degenerative diseases result from apoptotic photoreceptor cell death. As endogenously produced gaseous molecules such as hydrogen sulfide (H2S) and nitric oxide (NO) play a key role in apoptosis, we compared the expression levels of genes and proteins involved in the production of these molecules in the retina of normal dogs and three canine models (rcd1, crd2, and xlpra2) of human inherited retinal degeneration (IRD). Using qRT-PCR, Western blot, and immunohistochemistry (IHC), we showed that mRNA and protein levels of cystathionine β-synthase (CBS), an enzyme that produces H2S in neurons, are increased in retinal degeneration, but those of cystathionine γ-lyase (CSE), an enzyme involved in the production of glutathione (GSH), an antioxidant, are not. Such findings suggest that increased levels of H2S that are not counterbalanced by increased antioxidant potential may contribute to disease in affected retinas. We also studied the expression of neuronal and inducible nitric oxide synthase (nNOS and iNOS), the enzymes responsible for NO production. Western blot and IHC results revealed increased levels of nNOS and iNOS, resulting in increased NO levels in mutant retinas. Finally, photoreceptors are rich in polyunsaturated fatty acids (PUFAs) that can make these cells vulnerable to oxidative damage through reactive oxygen species (ROS). Our results showed increased levels of acrolein and hydroxynonenal (4HNE), two main toxic products of PUFAs, surrounding the membranes of photoreceptors in affected canines. Increased levels of these toxic products, together with increased NO and ROS, likely render these cells susceptible to an intrinsic apoptotic pathway involving mitochondrial membranes. To assess this possibility, we measured the levels of BCL2, an anti-apoptotic protein in the mitochondrial membrane. Western blot results showed decreased levels of BCL2 protein in affected retinas. Overall, the results of this study identify alterations in the expression of enzymes directly involved in maintaining the normal redox status of the retina during retinal degeneration, thereby supporting future studies to investigate the role of H2S and NO in retinal degeneration and apoptosis.
Collapse
Affiliation(s)
- Alireza Badiei
- Department of Veterinary Medicine, College of Natural Science and Mathematics, University of Alaska Fairbanks, AK, USA; Division of Experimental Retinal Therapies, Department of Clinical Sciences and Advanced Medicine, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| | - William A Beltran
- Division of Experimental Retinal Therapies, Department of Clinical Sciences and Advanced Medicine, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Gustavo D Aguirre
- Division of Experimental Retinal Therapies, Department of Clinical Sciences and Advanced Medicine, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|