51
|
Khan MM, Kirabo A. Long Noncoding RNA MALAT1: Salt-Sensitive Hypertension. Int J Mol Sci 2024; 25:5507. [PMID: 38791545 PMCID: PMC11122212 DOI: 10.3390/ijms25105507] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Revised: 05/06/2024] [Accepted: 05/15/2024] [Indexed: 05/26/2024] Open
Abstract
Hypertension stands as the leading global cause of mortality, affecting one billion individuals and serving as a crucial risk indicator for cardiovascular morbidity and mortality. Elevated salt intake triggers inflammation and hypertension by activating antigen-presenting cells (APCs). We found that one of the primary reasons behind this pro-inflammatory response is the epithelial sodium channel (ENaC), responsible for transporting sodium ions into APCs and the activation of NADPH oxidase, leading to increased oxidative stress. Oxidative stress increases lipid peroxidation and the formation of pro-inflammatory isolevuglandins (IsoLG). Long noncoding RNAs (lncRNAs) play a crucial role in regulating gene expression, and MALAT1, broadly expressed across cell types, including blood vessels and inflammatory cells, is also associated with inflammation regulation. In hypertension, the decreased transcriptional activity of nuclear factor erythroid 2-related factor 2 (Nrf2 or Nfe2l2) correlates with heightened oxidative stress in APCs and impaired control of various antioxidant genes. Kelch-like ECH-associated protein 1 (Keap1), an intracellular inhibitor of Nrf2, exhibits elevated levels of hypertension. Sodium, through an increase in Sp1 transcription factor binding at its promoter, upregulates MALAT1 expression. Silencing MALAT1 inhibits sodium-induced Keap1 upregulation, facilitating the nuclear translocation of Nrf2 and subsequent antioxidant gene transcription. Thus, MALAT1, acting via the Keap1-Nrf2 pathway, modulates antioxidant defense in hypertension. This review explores the potential role of the lncRNA MALAT1 in controlling the Keap1-Nrf2-antioxidant defense pathway in salt-induced hypertension. The inhibition of MALAT1 holds therapeutic potential for the progression of salt-induced hypertension and cardiovascular disease (CVD).
Collapse
Affiliation(s)
- Mohd Mabood Khan
- Department of Medicine, Preston Research Building, Vanderbilt University Medical Centre, Nashville, TN 37232, USA
| | - Annet Kirabo
- Department of Medicine, Preston Research Building, Vanderbilt University Medical Centre, Nashville, TN 37232, USA
| |
Collapse
|
52
|
Dupuy J, Fouché E, Noirot C, Martin P, Buisson C, Guéraud F, Pierre F, Héliès-Toussaint C. A dual model of normal vs isogenic Nrf2-depleted murine epithelial cells to explore oxidative stress involvement. Sci Rep 2024; 14:10905. [PMID: 38740939 DOI: 10.1038/s41598-024-60938-2] [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: 12/16/2022] [Accepted: 04/29/2024] [Indexed: 05/16/2024] Open
Abstract
Cancer-derived cell lines are useful tools for studying cellular metabolism and xenobiotic toxicity, but they are not suitable for modeling the biological effects of food contaminants or natural biomolecules on healthy colonic epithelial cells in a normal genetic context. The toxicological properties of such compounds may rely on their oxidative properties. Therefore, it appears to be necessary to develop a dual-cell model in a normal genetic context that allows to define the importance of oxidative stress in the observed toxicity. Given that the transcription factor nuclear factor erythroid 2-related factor 2 (Nrf2) is considered to be the master regulator of antioxidant defenses, our aim was to develop a cellular model comparing normal and Nrf2-depleted isogenic cells to qualify oxidative stress-related toxicity. We generated these cells by using the CRISPR/Cas9 technique. Whole-genome sequencing enabled us to confirm that our cell lines were free of cancer-related mutations. We used 4-hydroxy-2-nonenal (HNE), a lipid peroxidation product closely related to oxidative stress, as a model molecule. Here we report significant differences between the two cell lines in glutathione levels, gene regulation, and cell viability after HNE treatment. The results support the ability of our dual-cell model to study the role of oxidative stress in xenobiotic toxicity.
Collapse
Affiliation(s)
- Jacques Dupuy
- National Research Institute for Agriculture and Environment (INRAE), Toxalim (Research Centre in Food Toxicology), INRAE, ENVT, INP-Purpan, UPS, Université de Toulouse, 180 Chemin de Tournefeuille, BP93173, 31027, Toulouse Cedex 3, France
| | - Edwin Fouché
- National Research Institute for Agriculture and Environment (INRAE), Toxalim (Research Centre in Food Toxicology), INRAE, ENVT, INP-Purpan, UPS, Université de Toulouse, 180 Chemin de Tournefeuille, BP93173, 31027, Toulouse Cedex 3, France
| | - Céline Noirot
- National Research Institute for Agriculture and Environment (INRAE), Université Fédérale de Toulouse, INRAE, BioinfOmics, GenoToul Bioinformatics Facility, 31326, Castanet-Tolosan, France
| | - Pierre Martin
- National Research Institute for Agriculture and Environment (INRAE), Université Fédérale de Toulouse, INRAE, BioinfOmics, GenoToul Bioinformatics Facility, 31326, Castanet-Tolosan, France
| | - Charline Buisson
- National Research Institute for Agriculture and Environment (INRAE), Toxalim (Research Centre in Food Toxicology), INRAE, ENVT, INP-Purpan, UPS, Université de Toulouse, 180 Chemin de Tournefeuille, BP93173, 31027, Toulouse Cedex 3, France
| | - Françoise Guéraud
- National Research Institute for Agriculture and Environment (INRAE), Toxalim (Research Centre in Food Toxicology), INRAE, ENVT, INP-Purpan, UPS, Université de Toulouse, 180 Chemin de Tournefeuille, BP93173, 31027, Toulouse Cedex 3, France
| | - Fabrice Pierre
- National Research Institute for Agriculture and Environment (INRAE), Toxalim (Research Centre in Food Toxicology), INRAE, ENVT, INP-Purpan, UPS, Université de Toulouse, 180 Chemin de Tournefeuille, BP93173, 31027, Toulouse Cedex 3, France
| | - Cécile Héliès-Toussaint
- National Research Institute for Agriculture and Environment (INRAE), Toxalim (Research Centre in Food Toxicology), INRAE, ENVT, INP-Purpan, UPS, Université de Toulouse, 180 Chemin de Tournefeuille, BP93173, 31027, Toulouse Cedex 3, France.
| |
Collapse
|
53
|
Becker S, L'Ecuyer Z, Jones BW, Zouache MA, McDonnell FS, Vinberg F. Modeling complex age-related eye disease. Prog Retin Eye Res 2024; 100:101247. [PMID: 38365085 PMCID: PMC11268458 DOI: 10.1016/j.preteyeres.2024.101247] [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: 08/15/2023] [Revised: 01/31/2024] [Accepted: 02/02/2024] [Indexed: 02/18/2024]
Abstract
Modeling complex eye diseases like age-related macular degeneration (AMD) and glaucoma poses significant challenges, since these conditions depend highly on age-related changes that occur over several decades, with many contributing factors remaining unknown. Although both diseases exhibit a relatively high heritability of >50%, a large proportion of individuals carrying AMD- or glaucoma-associated genetic risk variants will never develop these diseases. Furthermore, several environmental and lifestyle factors contribute to and modulate the pathogenesis and progression of AMD and glaucoma. Several strategies replicate the impact of genetic risk variants, pathobiological pathways and environmental and lifestyle factors in AMD and glaucoma in mice and other species. In this review we will primarily discuss the most commonly available mouse models, which have and will likely continue to improve our understanding of the pathobiology of age-related eye diseases. Uncertainties persist whether small animal models can truly recapitulate disease progression and vision loss in patients, raising doubts regarding their usefulness when testing novel gene or drug therapies. We will elaborate on concerns that relate to shorter lifespan, body size and allometries, lack of macula and a true lamina cribrosa, as well as absence and sequence disparities of certain genes and differences in their chromosomal location in mice. Since biological, rather than chronological, age likely predisposes an organism for both glaucoma and AMD, more rapidly aging organisms like small rodents may open up possibilities that will make research of these diseases more timely and financially feasible. On the other hand, due to the above-mentioned anatomical and physiological features, as well as pharmacokinetic and -dynamic differences small animal models are not ideal to study the natural progression of vision loss or the efficacy and safety of novel therapies. In this context, we will also discuss the advantages and pitfalls of alternative models that include larger species, such as non-human primates and rabbits, patient-derived retinal organoids, and human organ donor eyes.
Collapse
Affiliation(s)
- Silke Becker
- John A. Moran Eye Center, University of Utah, Salt Lake City, UT, USA
| | - Zia L'Ecuyer
- John A. Moran Eye Center, University of Utah, Salt Lake City, UT, USA
| | - Bryan W Jones
- John A. Moran Eye Center, University of Utah, Salt Lake City, UT, USA
| | - Moussa A Zouache
- John A. Moran Eye Center, University of Utah, Salt Lake City, UT, USA
| | - Fiona S McDonnell
- John A. Moran Eye Center, University of Utah, Salt Lake City, UT, USA; Biomedical Engineering, University of Utah, Salt Lake City, UT, USA
| | - Frans Vinberg
- John A. Moran Eye Center, University of Utah, Salt Lake City, UT, USA; Biomedical Engineering, University of Utah, Salt Lake City, UT, USA.
| |
Collapse
|
54
|
Wilson EN, Wang C, Swarovski MS, Zera KA, Ennerfelt HE, Wang Q, Chaney A, Gauba E, Ramos Benitez JA, Le Guen Y, Minhas PS, Panchal M, Tan YJ, Blacher E, A Iweka C, Cropper H, Jain P, Liu Q, Mehta SS, Zuckerman AJ, Xin M, Umans J, Huang J, Durairaj AS, Serrano GE, Beach TG, Greicius MD, James ML, Buckwalter MS, McReynolds MR, Rabinowitz JD, Andreasson KI. TREM1 disrupts myeloid bioenergetics and cognitive function in aging and Alzheimer disease mouse models. Nat Neurosci 2024; 27:873-885. [PMID: 38539014 PMCID: PMC11102654 DOI: 10.1038/s41593-024-01610-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Accepted: 02/22/2024] [Indexed: 04/21/2024]
Abstract
Human genetics implicate defective myeloid responses in the development of late-onset Alzheimer disease. A decline in peripheral and brain myeloid metabolism, triggering maladaptive immune responses, is a feature of aging. The role of TREM1, a pro-inflammatory factor, in neurodegenerative diseases is unclear. Here we show that Trem1 deficiency prevents age-dependent changes in myeloid metabolism, inflammation and hippocampal memory function in mice. Trem1 deficiency rescues age-associated declines in ribose 5-phosphate. In vitro, Trem1-deficient microglia are resistant to amyloid-β42 oligomer-induced bioenergetic changes, suggesting that amyloid-β42 oligomer stimulation disrupts homeostatic microglial metabolism and immune function via TREM1. In the 5XFAD mouse model, Trem1 haploinsufficiency prevents spatial memory loss, preserves homeostatic microglial morphology, and reduces neuritic dystrophy and changes in the disease-associated microglial transcriptomic signature. In aging APPSwe mice, Trem1 deficiency prevents hippocampal memory decline while restoring synaptic mitochondrial function and cerebral glucose uptake. In postmortem Alzheimer disease brain, TREM1 colocalizes with Iba1+ cells around amyloid plaques and its expression is associated with Alzheimer disease clinical and neuropathological severity. Our results suggest that TREM1 promotes cognitive decline in aging and in the context of amyloid pathology.
Collapse
Affiliation(s)
- Edward N Wilson
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA
- Wu Tsai Neurosciences Institute, Stanford University, Stanford, CA, USA
| | - Congcong Wang
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA
| | - Michelle S Swarovski
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA
| | - Kristy A Zera
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA
| | - Hannah E Ennerfelt
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA
| | - Qian Wang
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA
| | - Aisling Chaney
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA
- Department of Radiology, Stanford University School of Medicine, Stanford, CA, USA
| | - Esha Gauba
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA
| | - Javier A Ramos Benitez
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA
| | - Yann Le Guen
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA
| | - Paras S Minhas
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA
| | - Maharshi Panchal
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA
| | - Yuting J Tan
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA
| | - Eran Blacher
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA
| | - Chinyere A Iweka
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA
| | - Haley Cropper
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA
- Department of Radiology, Stanford University School of Medicine, Stanford, CA, USA
| | - Poorva Jain
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA
- Department of Radiology, Stanford University School of Medicine, Stanford, CA, USA
| | - Qingkun Liu
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA
| | - Swapnil S Mehta
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA
| | - Abigail J Zuckerman
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA
| | - Matthew Xin
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA
| | - Jacob Umans
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA
| | - Jolie Huang
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA
| | - Aarooran S Durairaj
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA
| | - Geidy E Serrano
- Civin Laboratory for Neuropathology, Banner Sun Health Research Institute, Sun City, AZ, USA
| | - Thomas G Beach
- Civin Laboratory for Neuropathology, Banner Sun Health Research Institute, Sun City, AZ, USA
| | - Michael D Greicius
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA
- Wu Tsai Neurosciences Institute, Stanford University, Stanford, CA, USA
| | - Michelle L James
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA
- Wu Tsai Neurosciences Institute, Stanford University, Stanford, CA, USA
- Department of Radiology, Stanford University School of Medicine, Stanford, CA, USA
| | - Marion S Buckwalter
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA
- Wu Tsai Neurosciences Institute, Stanford University, Stanford, CA, USA
- Department of Neurosurgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Melanie R McReynolds
- Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ, USA
- Department of Chemistry, Princeton University, Princeton, NJ, USA
- Department of Biochemistry and Molecular Biology, Huck Institutes of the Life Sciences, Pennsylvania State University, University Park, PA, USA
| | - Joshua D Rabinowitz
- Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ, USA
- Department of Chemistry, Princeton University, Princeton, NJ, USA
| | - Katrin I Andreasson
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA.
- Wu Tsai Neurosciences Institute, Stanford University, Stanford, CA, USA.
- Chan Zuckerberg Biohub, San Francisco, CA, USA.
| |
Collapse
|
55
|
Taylor AL, Dubuisson O, Pandey P, Zunica ERM, Vandanmagsar B, Dantas WS, Johnson A, Axelrod CL, Kirwan JP. Restricting bioenergetic efficiency enhances longevity and mitochondrial redox capacity in Drosophila melanogaster. Aging Cell 2024; 23:e14107. [PMID: 38343281 PMCID: PMC11113268 DOI: 10.1111/acel.14107] [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: 08/21/2023] [Revised: 01/24/2024] [Accepted: 01/29/2024] [Indexed: 05/24/2024] Open
Abstract
Mitochondria are essential for survival and as such, impairments in organelle homeostasis significantly accelerate age-related morbidity and mortality. Here, we determined the contribution of bioenergetic efficiency to life span and health span in Drosophila melanogaster utilizing the mitochondrial uncoupler BAM15. Life span was determined in flies fed a normal diet (ND) or high fat diet (HFD) supplemented with vehicle or BAM15. Locomotor function was determined by negative geotaxis assay in middle-aged flies fed vehicle or BAM15 under ND or HFD conditions. Redox capacity (high-resolution respirometry/fluorometry), citrate synthase (enzyme activity), mtDNA content (qPCR), gene expression (qPCR), and protein expression (western blot) were assessed in flight muscle homogenates of middle-aged flies fed vehicle or BAM15 ND. The molar ratio of H2O2 and O2 (H2O2:O2) in a defined respiratory state was calculated as a measure of redox balance. BAM15 extended life span by 9% on ND and 25% on HFD and improved locomotor activity by 125% on ND and 53% on HFD. Additionally, BAM15 enhanced oxidative phosphorylation capacity supported by pyruvate + malate, proline, and glycerol 3-phosphate. Concurrently, BAM15 enhanced the mitochondrial H2O2 production rate, reverse electron flow from mitochondrial glycerol-3-phosphate dehydrogenase (mGPDH) to Complex I, mGPDH, and Complex I without altering the H2O2:O2 ratio. BAM15 upregulated transcriptional signatures associated with mitochondrial function and fitness as well as antioxidant defense. BAM15-mediated restriction of bioenergetic efficiency prolongs life span and health span in Drosophila fed a ND or HFD. Improvements in life span and health span in ND were supported by synergistic enhancement of muscular redox capacity.
Collapse
Affiliation(s)
- Analisa L. Taylor
- Integrated Physiology and Molecular Medicine LaboratoryPennington Biomedical Research CenterBaton RougeLouisianaUSA
| | - Olga Dubuisson
- Department of Biological SciencesLouisiana State UniversityBaton RougeLouisianaUSA
| | - Pritika Pandey
- Department of Biological SciencesLouisiana State UniversityBaton RougeLouisianaUSA
| | - Elizabeth R. M. Zunica
- Integrated Physiology and Molecular Medicine LaboratoryPennington Biomedical Research CenterBaton RougeLouisianaUSA
| | - Bolormaa Vandanmagsar
- Integrated Physiology and Molecular Medicine LaboratoryPennington Biomedical Research CenterBaton RougeLouisianaUSA
| | - Wagner S. Dantas
- Integrated Physiology and Molecular Medicine LaboratoryPennington Biomedical Research CenterBaton RougeLouisianaUSA
| | - Alyssa Johnson
- Department of Biological SciencesLouisiana State UniversityBaton RougeLouisianaUSA
| | - Christopher L. Axelrod
- Integrated Physiology and Molecular Medicine LaboratoryPennington Biomedical Research CenterBaton RougeLouisianaUSA
| | - John P. Kirwan
- Integrated Physiology and Molecular Medicine LaboratoryPennington Biomedical Research CenterBaton RougeLouisianaUSA
| |
Collapse
|
56
|
Feng Q, Ruan X, Lu M, Bu S, Zhang Y. Metformin protects retinal pigment epithelium cells against H 2O 2-induced oxidative stress and inflammation via the Nrf2 signaling cascade. Graefes Arch Clin Exp Ophthalmol 2024; 262:1519-1530. [PMID: 38059999 DOI: 10.1007/s00417-023-06321-9] [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: 05/05/2023] [Revised: 10/06/2023] [Accepted: 11/17/2023] [Indexed: 12/08/2023] Open
Abstract
PURPOSE Dysfunctions of retinal pigment epithelium (RPE) attributed to oxidative stress and inflammation are implicated with age-related macular degeneration (AMD). A debate on the curative role of metformin in AMD has been raised, though several recent clinical studies support the lower odds by using metformin. This study aimed to determine whether metformin could exert cytoprotection against RPE oxidative damages and the potential mechanisms. METHODS A cellular AMD model was established by treating ARPE-19 cells with hydrogen peroxide (H2O2) for 24 h. The reactive oxygen species (ROS) generation, expression of antioxidant enzymes, and levels of pro-inflammatory cytokines were monitored under administrations with H2O2 with/without metformin. The expression and DNA-binding activity of transcription factor erythroid-related factor 2 (Nrf2) were determined by western blot, immunofluorescence, and electrophoretic mobility shift assay. Knockout of Nrf2 was conducted by CRISPR/Cas9 gene deletion system. RESULTS Metformin pretreatment significantly improved the H2O2-induced low viability of ARPE-19 cells, reduced ROS production, and increased contents of antioxidative molecules. Concurrently, metformin also suppressed levels of pro-inflammatory cytokines caused by H2O2. The metformin-augmented nuclear translocation and DNA-binding activity of Nrf2 were further verified by the increased expression of its downstream targets. Genetic deletion of Nrf2 blocked the cytoprotective role of metformin. CONCLUSION Metformin possesses antioxidative and anti-inflammatory properties in ARPE-19 cells by activating the Nrf2 signaling. It supports the potential use for the control and prevention of AMD.
Collapse
Affiliation(s)
- Qiting Feng
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, China
| | - Xiangcai Ruan
- Department of Anesthesia and Pain Medicine, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Min Lu
- Sanshui Huaxia Eye Hospital, Huaxia Eye Hospital Group, Foshan, China
| | - Shimiao Bu
- Department of Ophthalmology, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, 510080, China
| | - Yuehong Zhang
- Department of Ophthalmology, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, 510080, China.
| |
Collapse
|
57
|
Meichtry LB, Sotelo MB, Musachio EAS, Janner DE, Dahleh MMM, Fernandes EJ, Bortolotto VC, Guerra GP, Prigol M. Early exposure to trans fat causes cognitive impairment by modulating the expression of proteins associated with oxidative stress and synaptic plasticity in Drosophila melanogaster. Comp Biochem Physiol C Toxicol Pharmacol 2024; 279:109858. [PMID: 38369039 DOI: 10.1016/j.cbpc.2024.109858] [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: 09/30/2023] [Revised: 01/21/2024] [Accepted: 02/15/2024] [Indexed: 02/20/2024]
Abstract
Evidence has shown that consuming trans fatty acids (TFA) during development leads to their incorporation into the nervous tissue, resulting in neurological changes in flies. In this study, Drosophila melanogaster was exposed to different concentrations of hydrogenated vegetable fat (HVF) during development: substitute hydrogenated vegetable fat (SHVF), HVF 10 %, and HVF 20 %. The objective was to evaluate the effects of early trans fat exposure on cognition and associated pathways in flies. The results showed that early TFA exposure provoked a cerebral redox imbalance, as confirmed by increased reactive species (HVF 10 and 20 %) and lipid peroxidation (SHVF, HVF 10, and 20 %), reduced nuclear factor erythroid 2-related factor 2 immunoreactivity (HVF 10 and 20 %), and increased heat shock protein 70 (HVF 20 %), which was possibly responsible for decreasing superoxide dismutase (SHVF, HVF 10, and 20 %) and catalase (HVF 20 %) activities. Furthermore, the presence of TFA in nervous tissue impaired learning (HVF 10 and 20 %) and memory at 6 and 24 h (SHVF, HVF 10, and 20 %). These cognitive impairments may be linked to reduced Shank levels (HVF 20 %) and increased acetylcholinesterase activity (SHVF, HVF 10 and 20 %) observed. Our findings demonstrate that early exposure to trans fat leads to cerebral redox imbalance, altering proteins associated with stress, synaptic plasticity, and the cholinergic system, consequently leading to cognitive impairment in flies.
Collapse
Affiliation(s)
- Luana Barreto Meichtry
- Laboratory of Pharmacological and Toxicological Evaluations Applied to Bioactive Molecules (LaftamBio), Federal University of Pampa, Campus Itaqui, 97650-000 Itaqui, RS, Brazil
| | - Magna Barrientos Sotelo
- Institute of Food Science and Technology, Federal University of Rio Grande do Sul, 91501-970 Porto Alegre, RS, Brazil
| | - Elize Aparecida Santos Musachio
- Laboratory of Pharmacological and Toxicological Evaluations Applied to Bioactive Molecules (LaftamBio), Federal University of Pampa, Campus Itaqui, 97650-000 Itaqui, RS, Brazil
| | - Dieniffer Espinosa Janner
- Laboratory of Pharmacological and Toxicological Evaluations Applied to Bioactive Molecules (LaftamBio), Federal University of Pampa, Campus Itaqui, 97650-000 Itaqui, RS, Brazil
| | - Mustafa Munir Mustafa Dahleh
- Laboratory of Pharmacological and Toxicological Evaluations Applied to Bioactive Molecules (LaftamBio), Federal University of Pampa, Campus Itaqui, 97650-000 Itaqui, RS, Brazil
| | - Eliana Jardim Fernandes
- Laboratory of Pharmacological and Toxicological Evaluations Applied to Bioactive Molecules (LaftamBio), Federal University of Pampa, Campus Itaqui, 97650-000 Itaqui, RS, Brazil
| | - Vandreza Cardoso Bortolotto
- Laboratory of Pharmacological and Toxicological Evaluations Applied to Bioactive Molecules (LaftamBio), Federal University of Pampa, Campus Itaqui, 97650-000 Itaqui, RS, Brazil
| | - Gustavo Petri Guerra
- Laboratory of Pharmacological and Toxicological Evaluations Applied to Bioactive Molecules (LaftamBio), Federal University of Pampa, Campus Itaqui, 97650-000 Itaqui, RS, Brazil
| | - Marina Prigol
- Laboratory of Pharmacological and Toxicological Evaluations Applied to Bioactive Molecules (LaftamBio), Federal University of Pampa, Campus Itaqui, 97650-000 Itaqui, RS, Brazil.
| |
Collapse
|
58
|
Li J, Wang Z, Wang X, Guo J, Wang L, He D, Duan X, Zhang C, Ren Y, Yang C. Age-Related Variations in the Population of Active Secondary Hair Follicles, Oxidative Stress and Antioxidant Parameters in Cashmere Goats. Animals (Basel) 2024; 14:1350. [PMID: 38731354 PMCID: PMC11083899 DOI: 10.3390/ani14091350] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Revised: 04/28/2024] [Accepted: 04/29/2024] [Indexed: 05/13/2024] Open
Abstract
The objective of this study was to investigate age-related changes in cashmere production and the population of active secondary hair follicles in cashmere goats across different age groups as well as to explore the association between secondary hair follicle activity and oxidative stress. A total of 104 adult Inner Mongolian ewe goats, aged between 2 and 7 years old, were randomly selected as experimental subjects. Skin samples were collected in August 2020 and cashmere samples were collected in April 2021. The cashmere fiber yield, staple length, and diameter showed age-related variations in cashmere goats aged 2 to 7 years (p < 0.05). Cashmere production was higher in goats aged 2-4 years compared to those aged 5-7 years (p < 0.05). There were no significant differences in the population of primary and secondary hair follicles among goats aged 2 to 7 years. However, the population of active secondary hair follicles varied significantly with age, with the younger group (aged 2-4 years) having a higher population than those aged 5-7 years (p < 0.05). A moderate negative correlation was observed between cashmere fiber diameter and the population of active secondary hair follicles (p < 0.05). Age-related variations in skin antioxidant capacity and oxidative damage were observed among cashmere goats aged 2 to 7 years old (p < 0.05). Goats aged 2 to 4 years exhibited higher antioxidant capacity and lower oxidative damage (p < 0.05). Interestingly, the skin's antioxidant capacity and oxidative damage exhibited significant positive and negative correlations with the population of active secondary hair follicles (p < 0.05). This study presents a novel approach to enhance the activity of secondary hair follicles and improve cashmere production performance through the regulation of oxidative stress.
Collapse
Affiliation(s)
- Junxia Li
- College of Animal Science, Shanxi Agricultural University, Taigu 030801, China; (J.L.); (Z.W.); (X.W.); (J.G.); (L.W.); (C.Z.)
| | - Zhenguo Wang
- College of Animal Science, Shanxi Agricultural University, Taigu 030801, China; (J.L.); (Z.W.); (X.W.); (J.G.); (L.W.); (C.Z.)
| | - Xiayuan Wang
- College of Animal Science, Shanxi Agricultural University, Taigu 030801, China; (J.L.); (Z.W.); (X.W.); (J.G.); (L.W.); (C.Z.)
| | - Jingxin Guo
- College of Animal Science, Shanxi Agricultural University, Taigu 030801, China; (J.L.); (Z.W.); (X.W.); (J.G.); (L.W.); (C.Z.)
| | - Liujia Wang
- College of Animal Science, Shanxi Agricultural University, Taigu 030801, China; (J.L.); (Z.W.); (X.W.); (J.G.); (L.W.); (C.Z.)
| | - Dong He
- Inner Mongolian Yiwei White Cashmere Goat Co., Ltd., Erdos 017000, China;
| | - Xinming Duan
- Nongfa Yuan (Hainan) Agricultural Development Co., Ltd., Haikou 570100, China;
| | - Chunxiang Zhang
- College of Animal Science, Shanxi Agricultural University, Taigu 030801, China; (J.L.); (Z.W.); (X.W.); (J.G.); (L.W.); (C.Z.)
| | - Youshe Ren
- College of Animal Science, Shanxi Agricultural University, Taigu 030801, China; (J.L.); (Z.W.); (X.W.); (J.G.); (L.W.); (C.Z.)
- Key Laboratory of Farm Animal Genetic Resources Exploration and Breeding of Shanxi Province, Taigu 030801, China
| | - Chunhe Yang
- College of Animal Science, Shanxi Agricultural University, Taigu 030801, China; (J.L.); (Z.W.); (X.W.); (J.G.); (L.W.); (C.Z.)
- Key Laboratory of Farm Animal Genetic Resources Exploration and Breeding of Shanxi Province, Taigu 030801, China
| |
Collapse
|
59
|
Carozza G, Zerti D, Tisi A, Ciancaglini M, Maccarrone M, Maccarone R. An overview of retinal light damage models for preclinical studies on age-related macular degeneration: identifying molecular hallmarks and therapeutic targets. Rev Neurosci 2024; 35:303-330. [PMID: 38153807 DOI: 10.1515/revneuro-2023-0130] [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: 10/21/2023] [Accepted: 11/19/2023] [Indexed: 12/30/2023]
Abstract
Age-related macular degeneration (AMD) is a complex, multifactorial disease leading to progressive and irreversible retinal degeneration, whose pathogenesis has not been fully elucidated yet. Due to the complexity and to the multiple features of the disease, many efforts have been made to develop animal models which faithfully reproduce the overall AMD hallmarks or that are able to mimic the different AMD stages. In this context, light damage (LD) rodent models of AMD represent a suitable and reliable approach to mimic the different AMD forms (dry, wet and geographic atrophy) while maintaining the time-dependent progression of the disease. In this review, we comprehensively reported how the LD paradigms reproduce the main features of human AMD. We discuss the capability of these models to broaden the knowledge in AMD research, with a focus on the mechanisms and the molecular hallmarks underlying the pathogenesis of the disease. We also critically revise the remaining challenges and future directions for the use of LD models.
Collapse
Affiliation(s)
- Giulia Carozza
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, 67100 L'Aquila, Italy
| | - Darin Zerti
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, 67100 L'Aquila, Italy
| | - Annamaria Tisi
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, 67100 L'Aquila, Italy
| | - Marco Ciancaglini
- Department of Life, Health & Environmental Sciences, University of L'Aquila, 67100 L'Aquila, Italy
| | - Mauro Maccarrone
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, 67100 L'Aquila, Italy
- European Center for Brain Research (CERC)/Santa Lucia Foundation IRCCS, 00143 Rome, Italy
| | - Rita Maccarone
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, 67100 L'Aquila, Italy
| |
Collapse
|
60
|
Burdeau JA, Stephenson BJK, Aris IM, Preston EV, Hivert MF, Oken E, Mahalingaiah S, Chavarro JE, Calafat AM, Rifas-Shiman SL, Zota AR, James-Todd T. First trimester plasma PER- AND Polyfluoroalkyl Substances (PFAS) and blood pressure trajectories across the second and third trimesters of pregnancy. ENVIRONMENT INTERNATIONAL 2024; 186:108628. [PMID: 38583297 PMCID: PMC11196104 DOI: 10.1016/j.envint.2024.108628] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Revised: 03/31/2024] [Accepted: 04/02/2024] [Indexed: 04/09/2024]
Abstract
BACKGROUND Evidence suggests that exposure to per- and polyfluoroalkyl substances (PFAS) increases risk of high blood pressure (BP) during pregnancy. Prior studies did not examine associations with BP trajectory parameters (i.e., overall magnitude and velocity) during pregnancy, which is linked to adverse pregnancy outcomes. OBJECTIVES To estimate associations of multiple plasma PFAS in early pregnancy with BP trajectory parameters across the second and third trimesters. To assess potential effect modification by maternal age and parity. METHODS In 1297 individuals, we quantified six PFAS in plasma collected during early pregnancy (median gestational age: 9.4 weeks). We abstracted from medical records systolic BP (SBP) and diastolic BP (DBP) measurements, recorded from 12 weeks gestation until delivery. BP trajectory parameters were estimated via Super Imposition by Translation and Rotation modeling. Subsequently, Bayesian Kernel Machine Regression (BKMR) was employed to estimate individual and joint associations of PFAS concentrations with trajectory parameters - adjusting for maternal age, race/ethnicity, pre-pregnancy body mass index, income, parity, smoking status, and seafood intake. We evaluated effect modification by age at enrollment and parity. RESULTS We collected a median of 13 BP measurements per participant. In BKMR, higher concentration of perfluorooctane sulfonate (PFOS) was independently associated with higher magnitude of overall SBP and DBP trajectories (i.e., upward shift of trajectories) and faster SBP trajectory velocity, holding all other PFAS at their medians. In stratified BKMR analyses, participants with ≥ 1 live birth had more pronounced positive associations between PFOS and SBP velocity, DBP magnitude, and DBP velocity - compared to nulliparous participants. We did not observe significant associations between concentrations of the overall PFAS mixture and either magnitude or velocity of the BP trajectories. CONCLUSION Early pregnancy plasma PFOS concentrations were associated with altered BP trajectory in pregnancy, which may impact future cardiovascular health of the mother.
Collapse
Affiliation(s)
- Jordan A Burdeau
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA.
| | - Briana J K Stephenson
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA, USA.
| | - Izzuddin M Aris
- Division of Chronic Disease Research Across the Lifecourse, Department of Population Medicine, Harvard Medical School and Harvard Pilgrim Health Care Institute, Boston, MA, USA.
| | - Emma V Preston
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, MA, USA.
| | - Marie-France Hivert
- Division of Chronic Disease Research Across the Lifecourse, Department of Population Medicine, Harvard Medical School and Harvard Pilgrim Health Care Institute, Boston, MA, USA; Diabetes Unit, Massachusetts General Hospital, Boston, MA, USA.
| | - Emily Oken
- Division of Chronic Disease Research Across the Lifecourse, Department of Population Medicine, Harvard Medical School and Harvard Pilgrim Health Care Institute, Boston, MA, USA.
| | - Shruthi Mahalingaiah
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, MA, USA; Department of Obstetrics and Gynecology, Division of Reproductive Endocrinology and Infertility, Massachusetts General Hospital, Boston, MA, USA.
| | - Jorge E Chavarro
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA; Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA, USA.
| | - Antonia M Calafat
- Division of Laboratory Sciences, National Center for Environmental Health, Centers for Disease Control and Prevention, Atlanta, GA, USA.
| | - Sheryl L Rifas-Shiman
- Division of Chronic Disease Research Across the Lifecourse, Department of Population Medicine, Harvard Medical School and Harvard Pilgrim Health Care Institute, Boston, MA, USA.
| | - Ami R Zota
- Department of Environmental Health Sciences, Columbia University Mailman School of Public Health, New York, NY, USA.
| | - Tamarra James-Todd
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA; Division of Chronic Disease Research Across the Lifecourse, Department of Population Medicine, Harvard Medical School and Harvard Pilgrim Health Care Institute, Boston, MA, USA.
| |
Collapse
|
61
|
Ji HW, Wang CR, Yuan XW, Wang J, Wang L, Cao QL, Li YH, Xu YN, Kim NH. Mangiferin improves early porcine embryonic development by reducing oxidative stress. Reprod Domest Anim 2024; 59:e14565. [PMID: 38646981 DOI: 10.1111/rda.14565] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Revised: 03/15/2024] [Accepted: 03/30/2024] [Indexed: 04/25/2024]
Abstract
Mangiferin (MGN) is primarily found in the fruits, leaves, and bark of plants of the Anacardiaceae family, including mangoes. MGN exhibits various pharmacological effects, such as protection of the liver and gallbladder, anti-lipid peroxidation, and cancer prevention. This study aimed to investigate the effects of MGN supplementation during in vitro culture (IVC) on the antioxidant capacity of early porcine embryos and the underlying mechanisms involved. Porcine parthenotes in the IVC medium were exposed to different concentrations of MGN (0, 0.01, 0.1, and 1 μM). The addition of 0.1 μM MGN significantly increased the blastocyst formation rate of porcine embryos while reducing the apoptotic index and autophagy. Furthermore, the expression of antioxidation-related (SOD2, GPX1, NRF2, UCHL1), cell pluripotency (SOX2, NANOG), and mitochondria-related (TFAM, PGC1α) genes was upregulated. In contrast, the expression of apoptosis-related (CAS3, BAX) and autophagy-related (LC3B, ATG5) genes decreased after MGN supplementation. These findings suggest that MGN improves early porcine embryonic development by reducing oxidative stress-related genes.
Collapse
Affiliation(s)
- He-Wei Ji
- Guangdong Provincial Key Laboratory of Large Animal Models for Biomedicine, School of Pharmacy and Food Engineering, Wuyi University, Jiangmen, China
| | - Chao-Rui Wang
- Guangdong Provincial Key Laboratory of Large Animal Models for Biomedicine, School of Pharmacy and Food Engineering, Wuyi University, Jiangmen, China
| | - Xiu-Wen Yuan
- Guangdong Provincial Key Laboratory of Large Animal Models for Biomedicine, School of Pharmacy and Food Engineering, Wuyi University, Jiangmen, China
| | - Jing Wang
- College of Life and Health, Hainan University, Haikou, China
| | - Lin Wang
- Qingdao Haier Biotechnology Co., Ltd., Qingdao, China
| | - Qi-Long Cao
- Qingdao Haier Biotechnology Co., Ltd., Qingdao, China
| | - Ying-Hua Li
- Guangdong Provincial Key Laboratory of Large Animal Models for Biomedicine, School of Pharmacy and Food Engineering, Wuyi University, Jiangmen, China
| | - Yong-Nan Xu
- Guangdong Provincial Key Laboratory of Large Animal Models for Biomedicine, School of Pharmacy and Food Engineering, Wuyi University, Jiangmen, China
| | - Nam-Hyung Kim
- Guangdong Provincial Key Laboratory of Large Animal Models for Biomedicine, School of Pharmacy and Food Engineering, Wuyi University, Jiangmen, China
| |
Collapse
|
62
|
Hu Y, Zhang F, Ikonomovic M, Yang T. The Role of NRF2 in Cerebrovascular Protection: Implications for Vascular Cognitive Impairment and Dementia (VCID). Int J Mol Sci 2024; 25:3833. [PMID: 38612642 PMCID: PMC11012233 DOI: 10.3390/ijms25073833] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2024] [Revised: 03/20/2024] [Accepted: 03/22/2024] [Indexed: 04/14/2024] Open
Abstract
Vascular cognitive impairment and dementia (VCID) represents a broad spectrum of cognitive decline secondary to cerebral vascular aging and injury. It is the second most common type of dementia, and the prevalence continues to increase. Nuclear factor erythroid 2-related factor 2 (NRF2) is enriched in the cerebral vasculature and has diverse roles in metabolic balance, mitochondrial stabilization, redox balance, and anti-inflammation. In this review, we first briefly introduce cerebrovascular aging in VCID and the NRF2 pathway. We then extensively discuss the effects of NRF2 activation in cerebrovascular components such as endothelial cells, vascular smooth muscle cells, pericytes, and perivascular macrophages. Finally, we summarize the clinical potential of NRF2 activators in VCID.
Collapse
Affiliation(s)
- Yizhou Hu
- Department of Neurology, University of Pittsburgh, Pittsburgh, PA 15216, USA; (Y.H.); (F.Z.); (M.I.)
- Pittsburgh Institute of Brain Disorders and Recovery, University of Pittsburgh, Pittsburgh, PA 15216, USA
- Department of Internal Medicine, University of Pittsburgh Medical Center (UPMC) McKeesport, McKeesport, PA 15132, USA
| | - Feng Zhang
- Department of Neurology, University of Pittsburgh, Pittsburgh, PA 15216, USA; (Y.H.); (F.Z.); (M.I.)
- Pittsburgh Institute of Brain Disorders and Recovery, University of Pittsburgh, Pittsburgh, PA 15216, USA
| | - Milos Ikonomovic
- Department of Neurology, University of Pittsburgh, Pittsburgh, PA 15216, USA; (Y.H.); (F.Z.); (M.I.)
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, PA 15216, USA
- Geriatric Research Education and Clinical Center, VA Pittsburgh Healthcare System, Pittsburgh, PA 15240, USA
| | - Tuo Yang
- Department of Neurology, University of Pittsburgh, Pittsburgh, PA 15216, USA; (Y.H.); (F.Z.); (M.I.)
- Pittsburgh Institute of Brain Disorders and Recovery, University of Pittsburgh, Pittsburgh, PA 15216, USA
- Department of Internal Medicine, University of Pittsburgh Medical Center (UPMC), Pittsburgh, PA 15216, USA
| |
Collapse
|
63
|
Yu MG, Gordin D, Fu J, Park K, Li Q, King GL. Protective Factors and the Pathogenesis of Complications in Diabetes. Endocr Rev 2024; 45:227-252. [PMID: 37638875 PMCID: PMC10911956 DOI: 10.1210/endrev/bnad030] [Citation(s) in RCA: 20] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Revised: 06/13/2023] [Accepted: 08/24/2023] [Indexed: 08/29/2023]
Abstract
Chronic complications of diabetes are due to myriad disorders of numerous metabolic pathways that are responsible for most of the morbidity and mortality associated with the disease. Traditionally, diabetes complications are divided into those of microvascular and macrovascular origin. We suggest revising this antiquated classification into diabetes complications of vascular, parenchymal, and hybrid (both vascular and parenchymal) tissue origin, since the profile of diabetes complications ranges from those involving only vascular tissues to those involving mostly parenchymal organs. A major paradigm shift has occurred in recent years regarding the pathogenesis of diabetes complications, in which the focus has shifted from studies on risks to those on the interplay between risk and protective factors. While risk factors are clearly important for the development of chronic complications in diabetes, recent studies have established that protective factors are equally significant in modulating the development and severity of diabetes complications. These protective responses may help explain the differential severity of complications, and even the lack of pathologies, in some tissues. Nevertheless, despite the growing number of studies on this field, comprehensive reviews on protective factors and their mechanisms of action are not available. This review thus focused on the clinical, biochemical, and molecular mechanisms that support the idea of endogenous protective factors, and their roles in the initiation and progression of chronic complications in diabetes. In addition, this review also aimed to identify the main needs of this field for future studies.
Collapse
Affiliation(s)
- Marc Gregory Yu
- Research Division, Joslin Diabetes Center, Boston, MA 02215, USA
- Department of Medicine, Harvard Medical School, Boston, MA 02215, USA
| | - Daniel Gordin
- Research Division, Joslin Diabetes Center, Boston, MA 02215, USA
- Department of Medicine, Harvard Medical School, Boston, MA 02215, USA
- Department of Nephrology, University of Helsinki and Helsinki University Central Hospital, Stenbäckinkatu 9, FI-00029 Helsinki, Finland
- Minerva Foundation Institute for Medical Research, Tukholmankatu 8, 00290 Helsinki, Finland
| | - Jialin Fu
- Research Division, Joslin Diabetes Center, Boston, MA 02215, USA
- Department of Medicine, Harvard Medical School, Boston, MA 02215, USA
| | - Kyoungmin Park
- Research Division, Joslin Diabetes Center, Boston, MA 02215, USA
- Department of Medicine, Harvard Medical School, Boston, MA 02215, USA
| | - Qian Li
- Research Division, Joslin Diabetes Center, Boston, MA 02215, USA
- Department of Medicine, Harvard Medical School, Boston, MA 02215, USA
| | - George Liang King
- Research Division, Joslin Diabetes Center, Boston, MA 02215, USA
- Department of Medicine, Harvard Medical School, Boston, MA 02215, USA
| |
Collapse
|
64
|
Vahidinia Z, Azami Tameh A, Barati S, Izadpanah M, Seyed Hosseini E. Nrf2 activation: a key mechanism in stem cell exosomes-mediated therapies. Cell Mol Biol Lett 2024; 29:30. [PMID: 38431569 PMCID: PMC10909300 DOI: 10.1186/s11658-024-00551-3] [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: 12/13/2023] [Accepted: 02/20/2024] [Indexed: 03/05/2024] Open
Abstract
Exosomes are nano-sized membrane extracellular vesicles which can be released from various types of cells. Exosomes originating from inflammatory or injured cells can have detrimental effects on recipient cells, while exosomes derived from stem cells not only facilitate the repair and regeneration of damaged tissues but also inhibit inflammation and provide protective effects against various diseases, suggesting they may serve as an alternative strategy of stem cells transplantation. Exosomes have a fundamental role in communication between cells, through the transfer of proteins, bioactive lipids and nucleic acids (like miRNAs and mRNAs) between cells. This transfer significantly impacts both the physiological and pathological functions of recipient cells. Nuclear factor erythroid 2-related factor 2 (Nrf2), a transcription factor, is able to mitigate damage caused by oxidative stress and inflammation through various signaling pathways. The positive effects resulting from the activation of the Nrf2 signaling pathway in different disorders have been documented in various types of literature. Studies have confirmed that exosomes derived from stem cells could act as Nrf2 effective agonists. However, limited studies have explored the Nrf2 role in the therapeutic effects of stem cell-derived exosomes. This review provides a comprehensive overview of the existing knowledge concerning the role of Nrf2 signaling pathways in the impact exerted by stem cell exosomes in some common diseases.
Collapse
Affiliation(s)
- Zeinab Vahidinia
- Anatomical Sciences Research Center, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran.
| | - Abolfazl Azami Tameh
- Anatomical Sciences Research Center, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran
| | - Shirin Barati
- Department of Anatomy, Saveh University of Medical Sciences, Saveh, Iran
| | - Melika Izadpanah
- Department of Anatomical Sciences, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Elahe Seyed Hosseini
- Gametogenesis Research Center, Institute for Basic Sciences, Kashan University of Medical Science, Kashan, Iran
| |
Collapse
|
65
|
Chen F, Wang Q, Xiao M, Lou D, Wufur R, Hu S, Zhang Z, Wang Y, Zhang Y. A novel crosstalk between Nrf2 and Smad2/3 bridged by two nuanced Keap1 isoforms with their divergent effects on these distinct family transcription factors. Free Radic Biol Med 2024; 213:190-207. [PMID: 38242246 DOI: 10.1016/j.freeradbiomed.2024.01.025] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Revised: 01/04/2024] [Accepted: 01/16/2024] [Indexed: 01/21/2024]
Abstract
The Keap1-Nrf2 signalling to transcriptionally regulate antioxidant response element (ARE)-driven target genes has been accepted as key redox-sensitive pathway governing a vast variety of cellular stresses during healthy survival and disease development. Herein, we identified two nuanced isoforms α and β of Keap1 in HepG2 cells, arising from its first and another in-frame translation starting codons, respectively. In identifying those differential expression genes monitored by Keap1α and/or Keap1β, an unusual interaction of Keap1 with Smad2/3 was discovered by parsing transcriptome sequencing, Keap1-interacting protein profiling and relevant immunoprecipitation data. Further examination validated that Smad2/3 enable physical interaction with Keap1, as well as its isoforms α and β, by both EDGETSD and DLG motifs in the linker regions between their MH1 and MH2 domains, such that the stability of Smad2/3 and transcriptional activity are enhanced with their prolonged half-lives and relevant signalling responses from the cytoplasmic to nuclear compartments. The activation of Smad2/3 by Keap1, Keap1α or Keap1β was much likely contributable to a coordinative or another competitive effect of Nrf2, particularly in distinct Keap1-based cellular responses to its cognate growth factor (i.e. TGF-β1) or redox stress (e.g. stimulated by tBHQ and DTT). Overall, this discovery presents a novel functional bridge crossing the Keap1-Nrf2 redox signalling and the TGF-β1-Smad2/3 pathways so as to coordinately regulate the healthy growth and development.
Collapse
Affiliation(s)
- Feilong Chen
- College of Bioengineering and Graduate School, Chongqing University, No. 174 Shazheng Street, Shapingba District, Chongqing, 40044, China; Chongqing University Jiangjin Hospital, School of Medicine, Chongqing University, No. 725 Jiangzhou Avenue, Dingshan Street, Jiangjin District, Chongqing, 402262, China; The Laboratory of Cell Biochemistry and Topogenetic Regulation, College of Bioengineering and Faculty of Medical Sciences, Chongqing University, No. 174 Shazheng Street, Shapingba District, Chongqing, 40044, China
| | - Qing Wang
- College of Bioengineering and Graduate School, Chongqing University, No. 174 Shazheng Street, Shapingba District, Chongqing, 40044, China; Chongqing University Jiangjin Hospital, School of Medicine, Chongqing University, No. 725 Jiangzhou Avenue, Dingshan Street, Jiangjin District, Chongqing, 402262, China; The Laboratory of Cell Biochemistry and Topogenetic Regulation, College of Bioengineering and Faculty of Medical Sciences, Chongqing University, No. 174 Shazheng Street, Shapingba District, Chongqing, 40044, China
| | - Mei Xiao
- College of Bioengineering and Graduate School, Chongqing University, No. 174 Shazheng Street, Shapingba District, Chongqing, 40044, China; The Laboratory of Cell Biochemistry and Topogenetic Regulation, College of Bioengineering and Faculty of Medical Sciences, Chongqing University, No. 174 Shazheng Street, Shapingba District, Chongqing, 40044, China
| | - Deshuai Lou
- Chongqing Key Laboratory of Medicinal Resources in the Three Gorges Reservoir Region, School of Biological and Chemical Engineering, Chongqing University of Education, Chongqing, 400067, China
| | - Reziyamu Wufur
- College of Bioengineering and Graduate School, Chongqing University, No. 174 Shazheng Street, Shapingba District, Chongqing, 40044, China; Chongqing University Jiangjin Hospital, School of Medicine, Chongqing University, No. 725 Jiangzhou Avenue, Dingshan Street, Jiangjin District, Chongqing, 402262, China; The Laboratory of Cell Biochemistry and Topogenetic Regulation, College of Bioengineering and Faculty of Medical Sciences, Chongqing University, No. 174 Shazheng Street, Shapingba District, Chongqing, 40044, China
| | - Shaofan Hu
- College of Bioengineering and Graduate School, Chongqing University, No. 174 Shazheng Street, Shapingba District, Chongqing, 40044, China; The Laboratory of Cell Biochemistry and Topogenetic Regulation, College of Bioengineering and Faculty of Medical Sciences, Chongqing University, No. 174 Shazheng Street, Shapingba District, Chongqing, 40044, China
| | - Zhengwen Zhang
- Laboratory of Neuroscience, Institute of Cognitive Neuroscience and School of Pharmacy, University College London, 29-39 Brunswick Square, London, WC1N 1AX, England, United Kingdom
| | - Yeqi Wang
- College of Bioengineering and Graduate School, Chongqing University, No. 174 Shazheng Street, Shapingba District, Chongqing, 40044, China
| | - Yiguo Zhang
- Chongqing University Jiangjin Hospital, School of Medicine, Chongqing University, No. 725 Jiangzhou Avenue, Dingshan Street, Jiangjin District, Chongqing, 402262, China; The Laboratory of Cell Biochemistry and Topogenetic Regulation, College of Bioengineering and Faculty of Medical Sciences, Chongqing University, No. 174 Shazheng Street, Shapingba District, Chongqing, 40044, China.
| |
Collapse
|
66
|
Cuciniello R, Luongo D, Maurano F, Crispi S, Bergamo P. Dietary conjugated linoleic acid downregulates the AlCl 3-induced hyperactivation of compensatory and maladaptive signalling in the mouse brain cortex. Free Radic Biol Med 2024; 213:102-112. [PMID: 38218550 DOI: 10.1016/j.freeradbiomed.2024.01.006] [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: 11/15/2023] [Revised: 12/23/2023] [Accepted: 01/04/2024] [Indexed: 01/15/2024]
Abstract
Oxidative stress, hyperactivation of compensatory mechanisms (unfolded protein response, UPR; nuclear factor erythroid 2-related factor 2, Nrf2) and the stimulation of maladaptive response (inflammation/apoptosis) are interconnected pathogenic processes occurring during Alzheimer's disease (AD) progression. The neuroprotective ability of dietary Conjugated linoleic acid (CLAmix) in a mouse model of AlCl3-induced AD was recently described but, the effects of AlCl3 or CLAmix intake on these pathogenic processes are still unknown. The effects of dietary AlCl3 or CLAmix - alone and in combination - were examined in the brain cortex of twenty-eight BalbC mice divided into 4 groups (n = 7 each). The neurotoxic effects of AlCl3 were investigated in animals treated for 5 weeks with 100 mg/kg/day (AL). CLAmix supplementation (600 mg/kg bw/day) for 7 weeks (CLA) was aimed at evaluating its modulatory effects on the Nrf2 pathway while its co-treatment with AlCl3 during the last 5 weeks of CLAmix intake (CLA + AL) was used to investigate its neuroprotective ability. Untreated mice were used as controls. In the CLA group, the NADPH oxidase (NOX) activation in the brain cortex was accompanied by the modulation of the Nrf2 pathway. By contrast, in the AL mice, the significant upregulation of oxidative stress markers, compensatory pathways (UPR/Nrf2), proinflammatory cytokines (IL-6, TNFα) and the proapoptotic protein Bax levels were found as compared with control. Notably, in CLA + AL mice, the marked decrease of oxidative stress, UPR/Nrf2 markers and proinflammatory cytokines levels were associated with the significant increase of the antiapoptotic protein Bcl2. The involvement of NOX in the adaptive response elicited by CLAmix along with its protective effects against the onset of several pathogenic processes triggered by AlCl3, broadens the knowledge of the mechanism underlying the pleiotropic activity of Nrf2 activators and sheds new light on their potential therapeutic use against neurodegenerative disorders.
Collapse
Affiliation(s)
- R Cuciniello
- Institute of Biosciences and Bio-Resources, National Research Council (CNR-IBBR), Naples, 80100, Italy; IRCCS Neuromed, Pozzilli, 86077, Isernia, Italy
| | - D Luongo
- Institute of Food Sciences, National Research Council (CNR-ISA), Avellino, 83100, Italy
| | - F Maurano
- Institute of Food Sciences, National Research Council (CNR-ISA), Avellino, 83100, Italy
| | - S Crispi
- Institute of Biosciences and Bio-Resources, National Research Council (CNR-IBBR), Naples, 80100, Italy
| | - P Bergamo
- Institute of Biosciences and Bio-Resources, National Research Council (CNR-IBBR), Naples, 80100, Italy.
| |
Collapse
|
67
|
Gurubaran IS. Mitochondrial damage and clearance in retinal pigment epithelial cells. Acta Ophthalmol 2024; 102 Suppl 282:3-53. [PMID: 38467968 DOI: 10.1111/aos.16661] [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: 01/24/2024] [Accepted: 01/31/2024] [Indexed: 03/13/2024]
Abstract
Age-related macular degeneration (AMD) is a devastating eye disease that causes permanent vision loss in the central part of the retina, known as the macula. Patients with such severe visual loss face a reduced quality of life and are at a 1.5 times greater risk of death compared to the general population. Currently, there is no cure for or effective treatment for dry AMD. There are several mechanisms thought to underlie the disease, for example, ageing-associated chronic oxidative stress, mitochondrial damage, harmful protein aggregation and inflammation. As a way of gaining a better understanding of the molecular mechanisms behind AMD and thus developing new therapies, we have created a peroxisome proliferator-activated receptor gamma coactivator 1-alpha and nuclear factor erythroid 2-related factor 2 (PGC1α/NFE2L2) double-knockout (dKO) mouse model that mimics many of the clinical features of dry AMD, including elevated levels of oxidative stress markers, damaged mitochondria, accumulating lysosomal lipofuscin and extracellular drusen-like structures in retinal pigment epithelial cells (RPE). In addition, a human RPE cell-based model was established to examine the impact of non-functional intracellular clearance systems on inflammasome activation. In this study, we found that there was a disturbance in the autolysosomal machinery responsible for clearing mitochondria in the RPE cells of one-year-old PGC1α/NFE2L2-deficient mice. The confocal immunohistochemical analysis revealed an increase in autophagosome marker microtubule-associated proteins 1A/1B light chain 3B (LC3B) as well as multiple mitophagy markers such as PTE-induced putative kinase 1 (PINK1) and E3 ubiquitin ligase (PARKIN), along with signs of damaged mitochondria. However, no increase in autolysosome formation was detected, nor was there a colocalization of the lysosomal marker LAMP2 or the mitochondrial marker, ATP synthase β. There was an upregulation of late autolysosomal fusion Ras-related protein (Rab7) in the perinuclear space of RPE cells, together with autofluorescent aggregates. Additionally, we observed an increase in the numbers of Toll-like receptors 3 and 9, while those of NOD-like receptor 3 were decreased in PGC1α/NFE2L2 dKO retinal specimens compared to wild-type animals. There was a trend towards increased complement component C5a and increased involvement of the serine protease enzyme, thrombin, in enhancing the terminal pathway producing C5a, independent of C3. The levels of primary acute phase C-reactive protein and receptor for advanced glycation end products were also increased in the PGC1α/NFE2L2 dKO retina. Furthermore, selective proteasome inhibition with epoxomicin promoted both nicotinamide adenine dinucleotide phosphate (NADPH) oxidase and mitochondrial-mediated oxidative stress, leading to the release of mitochondrial DNA to the cytosol, resulting in potassium efflux-dependent activation of the absent in melanoma 2 (AIM2) inflammasome and the subsequent secretion of interleukin-1β in ARPE-19 cells. In conclusion, the data suggest that there is at least a relative decrease in mitophagy, increases in the amounts of C5 and thrombin and decreased C3 levels in this dry AMD-like model. Moreover, selective proteasome inhibition evoked mitochondrial damage and AIM2 inflammasome activation in ARPE-19 cells.
Collapse
Affiliation(s)
- Iswariyaraja Sridevi Gurubaran
- Department of Medicine, Clinical Medicine Unit, University of Eastern Finland Institute of Clinical Medicine, Kuopio, Northern Savonia, Finland
| |
Collapse
|
68
|
He Z, Sun C, Ma Y, Chen X, Wang Y, Chen K, Xie F, Zhang Y, Yuan Y, Liu C. Rejuvenating Aged Bone Repair through Multihierarchy Reactive Oxygen Species-Regulated Hydrogel. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2306552. [PMID: 37848015 DOI: 10.1002/adma.202306552] [Citation(s) in RCA: 17] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Revised: 09/05/2023] [Indexed: 10/19/2023]
Abstract
Aging exacerbates the dysfunction of tissue regeneration at multiple levels and gradually diminishes individual's capacity to withstand stress, damage, and disease. The excessive accumulation of reactive oxygen species (ROS) is considered a hallmark feature of senescent stem cells, which causes oxidative stress, deteriorates the host microenvironment, and eventually becomes a critical obstacle for aged bone defect repair. Till now, the strategies cannot synchronously and thoroughly regulate intracellular and extracellular ROS in senescent cells. Herein, a multihierarchy ROS scavenging system for aged bone regeneration is developed by fabricating an injectable PEGylated poly(glycerol sebacate) (PEGS-NH2 )/poly(γ-glutamic acid) (γ-PGA) hydrogel containing rapamycin-loaded poly(diselenide-carbonate) nanomicelles (PSeR). This PSeR hydrogel exhibits highly sensitive ROS responsiveness to the local aged microenvironment and dynamically releases drug-loaded nanomicelles to scavenge the intracellular ROS accumulated in senescent bone mesenchymal stem cells. The PSeR hydrogel effectively tunes the antioxidant function and delays senescence of bone mesenchymal stem cells by safeguarding DNA replication in an oxidative environment, thereby promoting the self-renewal ability and enhancing the osteogenic capacity for aged bone repair in vitro and in vivo. Thus, this multihierarchy ROS-regulated hydrogel provides a new strategy for treating degenerative diseases.
Collapse
Affiliation(s)
- Zirui He
- Basic Science Center Project of National Natural Science Foundation of China, Key Laboratory for Ultrafine Materials of Ministry of Education and School of Materials Science and Engineering, East China University of Science and Technology, Shanghai, 200237, P. R. China
| | - Chuanhao Sun
- Key Laboratory of Advanced Polymeric Materials, Key Laboratory for Ultrafine Materials of Ministry of Education, School of Materials Science and Engineering, East China University of Science and Technology, Shanghai, 200237, P. R. China
| | - Yifan Ma
- Department of Biomedical Engineering, The Ohio State University, Columbus, OH, 43210, USA
| | - Xi Chen
- Basic Science Center Project of National Natural Science Foundation of China, Key Laboratory for Ultrafine Materials of Ministry of Education and School of Materials Science and Engineering, East China University of Science and Technology, Shanghai, 200237, P. R. China
| | - Ying Wang
- Basic Science Center Project of National Natural Science Foundation of China, Key Laboratory for Ultrafine Materials of Ministry of Education and School of Materials Science and Engineering, East China University of Science and Technology, Shanghai, 200237, P. R. China
| | - Kai Chen
- Basic Science Center Project of National Natural Science Foundation of China, Key Laboratory for Ultrafine Materials of Ministry of Education and School of Materials Science and Engineering, East China University of Science and Technology, Shanghai, 200237, P. R. China
| | - Fangru Xie
- Basic Science Center Project of National Natural Science Foundation of China, Key Laboratory for Ultrafine Materials of Ministry of Education and School of Materials Science and Engineering, East China University of Science and Technology, Shanghai, 200237, P. R. China
| | - Yan Zhang
- Key Laboratory of Advanced Polymeric Materials, Key Laboratory for Ultrafine Materials of Ministry of Education, School of Materials Science and Engineering, East China University of Science and Technology, Shanghai, 200237, P. R. China
| | - Yuan Yuan
- Basic Science Center Project of National Natural Science Foundation of China, Key Laboratory for Ultrafine Materials of Ministry of Education and School of Materials Science and Engineering, East China University of Science and Technology, Shanghai, 200237, P. R. China
| | - Changsheng Liu
- Basic Science Center Project of National Natural Science Foundation of China, Key Laboratory for Ultrafine Materials of Ministry of Education and School of Materials Science and Engineering, East China University of Science and Technology, Shanghai, 200237, P. R. China
| |
Collapse
|
69
|
Hu Z, Zhao J, Liu X, Li Y, Jiang H, Fang W, Long X. Glycyrrhizin regulates antioxidation through Nrf2 signaling pathway in rat temporomandibular joint osteoarthritis. J Oral Rehabil 2024; 51:611-622. [PMID: 37962287 DOI: 10.1111/joor.13621] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Revised: 07/25/2023] [Accepted: 10/28/2023] [Indexed: 11/15/2023]
Abstract
BACKGROUND Regulation of redox homeostasis could reduce osteoarthritis severity and limit disease progression, while glycyrrhizin (GL) shows great antioxidant and anti-inflammatory capacity. OBJECTIVE The aim of this study was to investigate the role of GL on oxidative stress and the potential regulatory mechanism in rat temporomandibular joint (TMJ) chondrocytes under oxidative stress, and investigate the effect of GL in the rat temporomandibular joint osteoarthritis (TMJOA) model. METHODS Rat TMJ chondrocytes were cultured in oxidative stress with different doses of GL. The effect of glycyrrhizin on the nuclear factor-erythroid 2-related factor 2 (Nrf2) in oxidative stress was evaluated by western blot and immunofluorescence staining. A rat model of TMJOA was treated with GL. Micro-computed tomography, histological and immunohistochemical analysis were used to assess the pathological change of TMJOA. RESULTS The expression of superoxide dismutase 1 (SOD1), heme oxygenase-1 (HO-1), and peroxiredoxin 6 (PRDX6) were decreased, and intracellular Nrf2 signaling pathway was activated in chondrocytes in oxidative stress. GL upregulates the expression of antioxidants, especially PRDX6, as well as increases Nrf2 expression and nuclear translocation in rat condylar chondrocytes. Administration of GL attenuates condylar bone destruction, cartilage degeneration, and synovitis in rats TMJOA. Meanwhile, GL alleviated oxidative stress and enhanced the antioxidant capacity of TMJOA cartilage. CONCLUSION This study suggested that GL alleviates rat TMJOA by regulating oxidative stress in condylar cartilage.
Collapse
Affiliation(s)
- Zhihui Hu
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School and Hospital of Stomatology, Wuhan University, Wuhan, China
- Department of Occlusion and Temporomandibular Joint Diseases, Tianjin Stomatological Hospital, School of Medicine, Nankai University, Tianjin, China
- Tianjin Key Laboratory of Oral and Maxillofacial Function Reconstruction, Tianjin, China
| | - Jie Zhao
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School and Hospital of Stomatology, Wuhan University, Wuhan, China
- Department of Oral and Maxillofacial Surgery, School and Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Xin Liu
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School and Hospital of Stomatology, Wuhan University, Wuhan, China
- Department of Oral and Maxillofacial Surgery, School and Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Yanyan Li
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School and Hospital of Stomatology, Wuhan University, Wuhan, China
- Department of Oral and Maxillofacial Surgery, School and Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Henghua Jiang
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School and Hospital of Stomatology, Wuhan University, Wuhan, China
- Department of Oral and Maxillofacial Surgery, School and Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Wei Fang
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School and Hospital of Stomatology, Wuhan University, Wuhan, China
- Department of Oral and Maxillofacial Surgery, School and Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Xing Long
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School and Hospital of Stomatology, Wuhan University, Wuhan, China
- Department of Oral and Maxillofacial Surgery, School and Hospital of Stomatology, Wuhan University, Wuhan, China
| |
Collapse
|
70
|
Chen L, Zhang L, Ye X, Deng Z, Zhao C. Ergothioneine and its congeners: anti-ageing mechanisms and pharmacophore biosynthesis. Protein Cell 2024; 15:191-206. [PMID: 37561026 PMCID: PMC10903977 DOI: 10.1093/procel/pwad048] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Accepted: 07/24/2023] [Indexed: 08/11/2023] Open
Abstract
Ergothioneine, Ovothiol, and Selenoneine are sulfur/selenium-containing histidine-derived natural products widely distributed across different organisms. They exhibit significant antioxidant properties, making them as potential lead compounds for promoting health. Increasing evidence suggests that Ergothioneine is positively correlated with healthy ageing and longevity. The mechanisms underlying Ergothioneine's regulation of the ageing process at cellular and molecular levels are beginning to be understood. In this review, we provide an in-depth and extensive coverage of the anti-ageing studies on Ergothioneine and discuss its possible intracellular targeting pathways. In addition, we highlight the recent efforts in elucidating the biosynthetic details for Ergothioneine, Ovothiol, and Selenoneine, with a particular focus on the study of their pharmacophore-forming enzymology.
Collapse
Affiliation(s)
- Li Chen
- Department of Geriatrics, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan 430072, China
- Key Laboratory of Combinatory Biosynthesis and Drug Discovery, School of Pharmaceutical Sciences, Ministry of Education, Wuhan University, Wuhan 430072, China
| | - Liping Zhang
- Key Laboratory of Combinatory Biosynthesis and Drug Discovery, School of Pharmaceutical Sciences, Ministry of Education, Wuhan University, Wuhan 430072, China
| | - Xujun Ye
- Department of Geriatrics, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan 430072, China
| | - Zixin Deng
- Department of Geriatrics, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan 430072, China
- Key Laboratory of Combinatory Biosynthesis and Drug Discovery, School of Pharmaceutical Sciences, Ministry of Education, Wuhan University, Wuhan 430072, China
| | - Changming Zhao
- Department of Geriatrics, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan 430072, China
- Key Laboratory of Combinatory Biosynthesis and Drug Discovery, School of Pharmaceutical Sciences, Ministry of Education, Wuhan University, Wuhan 430072, China
| |
Collapse
|
71
|
Díaz M, Valdés-Baizabal C, de Pablo DP, Marin R. Age-Dependent Changes in Nrf2/Keap1 and Target Antioxidant Protein Expression Correlate to Lipoxidative Adducts, and Are Modulated by Dietary N-3 LCPUFA in the Hippocampus of Mice. Antioxidants (Basel) 2024; 13:206. [PMID: 38397804 PMCID: PMC10886099 DOI: 10.3390/antiox13020206] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2023] [Revised: 01/26/2024] [Accepted: 01/30/2024] [Indexed: 02/25/2024] Open
Abstract
The brain has a high metabolism rate that may generate reactive oxygen and nitrogen species. Consequently, nerve cells require highly efficient antioxidant defenses in order to prevent a condition of deleterious oxidative stress. This is particularly relevant in the hippocampus, a highly complex cerebral area involved in processing superior cognitive functions. Most current evidence points to hippocampal oxidative damage as a causal effect for neurodegenerative disorders, especially Alzheimer's disease. Nuclear factor erythroid-2-related factor 2/Kelch-like ECH-associated protein 1 (Nrf2/Keap1) is a master key for the transcriptional regulation of antioxidant and detoxifying systems. It is ubiquitously expressed in brain areas, mainly supporting glial cells. In the present study, we have analyzed the relationships between Nrf2 and Keap1 isoforms in hippocampal tissue in response to aging and dietary long-chain polyunsaturated fatty acids (LCPUFA) supplementation. The possible involvement of lipoxidative and nitrosative by-products in the dynamics of the Nrf2/Keap1 complex was examined though determination of protein adducts, namely malondialdehyde (MDA), 4-hydroxynonenal (HNE), and 3-nitro-tyrosine (NTyr) under basal conditions. The results were correlated to the expression of target proteins heme-oxygenase-1 (HO-1) and glutathione peroxidase 4 (GPx4), whose expressions are known to be regulated by Nrf2/Keap1 signaling activation. All variables in this study were obtained simultaneously from the same preparations, allowing multivariate approaches. The results demonstrate a complex modification of the protein expression patterns together with the formation of adducts in response to aging and diet supplementation. Both parameters exhibited a strong interaction. Noticeably, LCPUFA supplementation to aged animals restored the Nrf2/Keap1/target protein patterns to the status observed in young animals, therefore driving a "rejuvenation" of hippocampal antioxidant defense.
Collapse
Affiliation(s)
- Mario Díaz
- Department of Physics, Faculty of Sciences, University of La Laguna, 38200 Tenerife, Spain
- Instituto Universitario de Neurociencias (IUNE), University of La Laguna, 38320 Tenerife, Spain; (C.V.-B.); (D.P.d.P.); (R.M.)
| | - Catalina Valdés-Baizabal
- Instituto Universitario de Neurociencias (IUNE), University of La Laguna, 38320 Tenerife, Spain; (C.V.-B.); (D.P.d.P.); (R.M.)
- Laboratory of Cellular Neurobiology, Department of Basic Medical Sciences, Faculty of Health Sciences, University of La Laguna, 38200 Tenerife, Spain
| | - Daniel Pereda de Pablo
- Instituto Universitario de Neurociencias (IUNE), University of La Laguna, 38320 Tenerife, Spain; (C.V.-B.); (D.P.d.P.); (R.M.)
- Laboratory of Cellular Neurobiology, Department of Basic Medical Sciences, Faculty of Health Sciences, University of La Laguna, 38200 Tenerife, Spain
| | - Raquel Marin
- Instituto Universitario de Neurociencias (IUNE), University of La Laguna, 38320 Tenerife, Spain; (C.V.-B.); (D.P.d.P.); (R.M.)
- Laboratory of Cellular Neurobiology, Department of Basic Medical Sciences, Faculty of Health Sciences, University of La Laguna, 38200 Tenerife, Spain
- Associate Research Unit ULL-CSIC “Membrane Physiology and Biophysics in Neurodegenerative and Cancer Diseases”, 38200 Tenerife, Spain
| |
Collapse
|
72
|
Łuczyńska K, Zhang Z, Pietras T, Zhang Y, Taniguchi H. NFE2L1/Nrf1 serves as a potential therapeutical target for neurodegenerative diseases. Redox Biol 2024; 69:103003. [PMID: 38150994 PMCID: PMC10788251 DOI: 10.1016/j.redox.2023.103003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2023] [Revised: 12/15/2023] [Accepted: 12/16/2023] [Indexed: 12/29/2023] Open
Abstract
The failure of the proper protein turnover in the nervous system is mainly linked to a variety of neurodegenerative disorders. Therefore, a better understanding of key protein degradation through the ubiquitin-proteasome system is critical for effective prevention and treatment of those disorders. The proteasome expression is tightly regulated by a CNC (cap'n'collar) family of transcription factors, amongst which the nuclear factor-erythroid 2-like bZIP factor 1 (NFE2L1, also known as Nrf1, with its long isoform TCF11 and short isoform LCR-F1) has been identified as an indispensable regulator of the transcriptional expression of the ubiquitin-proteasome system. However, much less is known about how the pivotal role of NFE2L1/Nrf1, as compared to its homologous NFE2L2 (also called Nrf2), is translated to its physiological and pathophysiological functions in the nervous system insomuch as to yield its proper cytoprotective effects against neurodegenerative diseases. The potential of NFE2L1 to fulfill its unique neuronal function to serve as a novel therapeutic target for neurodegenerative diseases is explored by evaluating the hitherto established preclinical and clinical studies of Alzheimer's and Parkinson's diseases. In this review, we have also showcased a group of currently available activators of NFE2L1, along with an additional putative requirement of this CNC-bZIP factor for healthy longevity based on the experimental evidence obtained from its orthologous SKN1-A in Caenorhabditis elegans.
Collapse
Affiliation(s)
- Kamila Łuczyńska
- Institute of Genetics and Animal Biotechnology of the Polish Academy of Sciences, Jastrzebiec, 05-552, Poland; The Second Department of Psychiatry, Institute of Psychiatry and Neurology in Warsaw, 02-957, Warsaw, Poland
| | - Zhengwen Zhang
- Laboratory of Neuroscience, Institute of Cognitive Neuroscience and School of Pharmacy, University College London, 29-39 Brunswick Square, London, WC1N 1AX, England, United Kingdom
| | - Tadeusz Pietras
- The Second Department of Psychiatry, Institute of Psychiatry and Neurology in Warsaw, 02-957, Warsaw, Poland; Department of Clinical Pharmacology, Medical University of Lodz, 90-153, Łódź, Poland
| | - Yiguo Zhang
- Chongqing University Jiangjin Hospital, School of Medicine, Chongqing University, No. 725 Jiangzhou Avenue, Dingshan Street, Jiangjin District, Chongqing, 402260, China; The Laboratory of Cell Biochemistry and Topogenetic Regulation, College of Bioengineering & Faculty of Medical Sciences, Chongqing University, No. 174 Shazheng Street, Shapingba District, Chongqing, 400044, China.
| | - Hiroaki Taniguchi
- Institute of Genetics and Animal Biotechnology of the Polish Academy of Sciences, Jastrzebiec, 05-552, Poland.
| |
Collapse
|
73
|
He Q, Hu D, Zheng F, Chen W, Hu K, Liu J, Yao C, Li H, Wei Y. Investigating the Nexus of NLRP3 Inflammasomes and COVID-19 Pathogenesis: Unraveling Molecular Triggers and Therapeutic Strategies. Viruses 2024; 16:213. [PMID: 38399989 PMCID: PMC10892947 DOI: 10.3390/v16020213] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2024] [Revised: 01/25/2024] [Accepted: 01/27/2024] [Indexed: 02/25/2024] Open
Abstract
The coronavirus disease 2019 (COVID-19) global pandemic, caused by severe acute respiratory syndrome coronavirus type 2 (SARS-CoV-2), has been marked by severe cases demonstrating a "cytokine storm", an upsurge of pro-inflammatory cytokines in the bloodstream. NLRP3 inflammasomes, integral to the innate immune system, are speculated to be activated by SARS-CoV-2 within host cells. This review investigates the potential correlation between NLRP3 inflammasomes and COVID-19, exploring the cellular and molecular mechanisms through which SARS-CoV-2 triggers their activation. Furthermore, promising strategies targeting NLRP3 inflammasomes are proposed to mitigate the excessive inflammatory response provoked by SARS-CoV-2 infection. By synthesizing existing studies, this paper offers insights into NLRP3 as a therapeutic target, elucidating the interplay between COVID-19 and its pathophysiology. It serves as a valuable reference for future clinical approaches in addressing COVID-19 by targeting NLRP3, thus providing potential avenues for therapeutic intervention.
Collapse
Affiliation(s)
- Qun He
- Sino-German Biomedical Center, National “111” Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Hubei University of Technology, Wuhan 430068, China; (Q.H.); (F.Z.); (W.C.); (K.H.); (J.L.); (C.Y.); (H.L.)
| | - Da Hu
- Sinopharm Animal Health Corporation Ltd., Wuhan 430075, China;
| | - Fuqiang Zheng
- Sino-German Biomedical Center, National “111” Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Hubei University of Technology, Wuhan 430068, China; (Q.H.); (F.Z.); (W.C.); (K.H.); (J.L.); (C.Y.); (H.L.)
| | - Wenxuan Chen
- Sino-German Biomedical Center, National “111” Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Hubei University of Technology, Wuhan 430068, China; (Q.H.); (F.Z.); (W.C.); (K.H.); (J.L.); (C.Y.); (H.L.)
| | - Kanghong Hu
- Sino-German Biomedical Center, National “111” Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Hubei University of Technology, Wuhan 430068, China; (Q.H.); (F.Z.); (W.C.); (K.H.); (J.L.); (C.Y.); (H.L.)
| | - Jinbiao Liu
- Sino-German Biomedical Center, National “111” Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Hubei University of Technology, Wuhan 430068, China; (Q.H.); (F.Z.); (W.C.); (K.H.); (J.L.); (C.Y.); (H.L.)
| | - Chenguang Yao
- Sino-German Biomedical Center, National “111” Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Hubei University of Technology, Wuhan 430068, China; (Q.H.); (F.Z.); (W.C.); (K.H.); (J.L.); (C.Y.); (H.L.)
| | - Hanluo Li
- Sino-German Biomedical Center, National “111” Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Hubei University of Technology, Wuhan 430068, China; (Q.H.); (F.Z.); (W.C.); (K.H.); (J.L.); (C.Y.); (H.L.)
| | - Yanhong Wei
- Sino-German Biomedical Center, National “111” Center for Cellular Regulation and Molecular Pharmaceutics, Key Laboratory of Fermentation Engineering (Ministry of Education), Hubei University of Technology, Wuhan 430068, China; (Q.H.); (F.Z.); (W.C.); (K.H.); (J.L.); (C.Y.); (H.L.)
| |
Collapse
|
74
|
Tejada S, Sarubbo F, Jiménez-García M, Ramis MR, Monserrat-Mesquida M, Quetglas-Llabrés MM, Capó X, Esteban S, Sureda A, Moranta D. Mitigating Age-Related Cognitive Decline and Oxidative Status in Rats Treated with Catechin and Polyphenon-60. Nutrients 2024; 16:368. [PMID: 38337652 PMCID: PMC10857701 DOI: 10.3390/nu16030368] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Revised: 01/23/2024] [Accepted: 01/24/2024] [Indexed: 02/12/2024] Open
Abstract
Aging is a normal physiological process influenced by the combination of multiple mechanisms, primarily oxidative stress and neuroinflammation, which impact general physiology and brain function. Phenolic compounds have demonstrated the ability to slow down the aging process of the brain due to their antioxidant and anti-inflammatory effects. This study assessed the protective properties of catechin and polyphenon-60 in non-pathologically aged rats regarding visuo-spatial learning and the oxidative status of the frontal cortex. Old animals were treated with catechin or green tea extract (polyphenon-60) for 36 days, daily. Healthy old and young rats were used as controls. During the first training phase, treated rats executed the test better, locating the target in less time compared with the controls. Biomarkers of oxidative stress (catalase activities, superoxide dismutase, glutathione reductase, and glutathione S-transferase) were reduced in the brain of old animals, although their activities were partially improved after both antioxidant treatments. Furthermore, the rise in the production of reactive oxygen species and malondialdehyde levels-a marker of lipid peroxidation-in the frontal cortex of aged animals was significantly ameliorated after the interventions. In conclusion, old rats exhibited enhanced cognitive function and reduced stress levels following the administration of catechin and polyphenon-60.
Collapse
Affiliation(s)
- Silvia Tejada
- Laboratory of Neurophysiology, University of the Balearic Islands, 07122 Palma de Mallorca, Spain; (S.T.); (F.S.); (M.J.-G.); (M.R.R.); (S.E.); (D.M.)
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III (ISCIII), 28029 Madrid, Spain
- Health Research Institute of Balearic Islands (IdISBa), 07120 Palma de Mallorca, Spain; (M.M.-M.); (M.M.Q.-L.); (X.C.)
| | - Fiorella Sarubbo
- Laboratory of Neurophysiology, University of the Balearic Islands, 07122 Palma de Mallorca, Spain; (S.T.); (F.S.); (M.J.-G.); (M.R.R.); (S.E.); (D.M.)
- Health Research Institute of Balearic Islands (IdISBa), 07120 Palma de Mallorca, Spain; (M.M.-M.); (M.M.Q.-L.); (X.C.)
| | - Manuel Jiménez-García
- Laboratory of Neurophysiology, University of the Balearic Islands, 07122 Palma de Mallorca, Spain; (S.T.); (F.S.); (M.J.-G.); (M.R.R.); (S.E.); (D.M.)
- Health Research Institute of Balearic Islands (IdISBa), 07120 Palma de Mallorca, Spain; (M.M.-M.); (M.M.Q.-L.); (X.C.)
| | - Margarida R. Ramis
- Laboratory of Neurophysiology, University of the Balearic Islands, 07122 Palma de Mallorca, Spain; (S.T.); (F.S.); (M.J.-G.); (M.R.R.); (S.E.); (D.M.)
| | - Margalida Monserrat-Mesquida
- Health Research Institute of Balearic Islands (IdISBa), 07120 Palma de Mallorca, Spain; (M.M.-M.); (M.M.Q.-L.); (X.C.)
- Research Group in Community Nutrition and Oxidative Stress (NUCOX), University of Balearic Islands, 07122 Palma de Mallorca, Spain
| | - Maria Magdalena Quetglas-Llabrés
- Health Research Institute of Balearic Islands (IdISBa), 07120 Palma de Mallorca, Spain; (M.M.-M.); (M.M.Q.-L.); (X.C.)
- Research Group in Community Nutrition and Oxidative Stress (NUCOX), University of Balearic Islands, 07122 Palma de Mallorca, Spain
| | - Xavier Capó
- Health Research Institute of Balearic Islands (IdISBa), 07120 Palma de Mallorca, Spain; (M.M.-M.); (M.M.Q.-L.); (X.C.)
- Research Group in Community Nutrition and Oxidative Stress (NUCOX), University of Balearic Islands, 07122 Palma de Mallorca, Spain
| | - Susana Esteban
- Laboratory of Neurophysiology, University of the Balearic Islands, 07122 Palma de Mallorca, Spain; (S.T.); (F.S.); (M.J.-G.); (M.R.R.); (S.E.); (D.M.)
- Health Research Institute of Balearic Islands (IdISBa), 07120 Palma de Mallorca, Spain; (M.M.-M.); (M.M.Q.-L.); (X.C.)
| | - Antoni Sureda
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III (ISCIII), 28029 Madrid, Spain
- Health Research Institute of Balearic Islands (IdISBa), 07120 Palma de Mallorca, Spain; (M.M.-M.); (M.M.Q.-L.); (X.C.)
- Research Group in Community Nutrition and Oxidative Stress (NUCOX), University of Balearic Islands, 07122 Palma de Mallorca, Spain
| | - David Moranta
- Laboratory of Neurophysiology, University of the Balearic Islands, 07122 Palma de Mallorca, Spain; (S.T.); (F.S.); (M.J.-G.); (M.R.R.); (S.E.); (D.M.)
- Health Research Institute of Balearic Islands (IdISBa), 07120 Palma de Mallorca, Spain; (M.M.-M.); (M.M.Q.-L.); (X.C.)
| |
Collapse
|
75
|
Gavito-Covarrubias D, Ramírez-Díaz I, Guzmán-Linares J, Limón ID, Manuel-Sánchez DM, Molina-Herrera A, Coral-García MÁ, Anastasio E, Anaya-Hernández A, López-Salazar P, Juárez-Díaz G, Martínez-Juárez J, Torres-Jácome J, Albarado-Ibáñez A, Martínez-Laguna Y, Morán C, Rubio K. Epigenetic mechanisms of particulate matter exposure: air pollution and hazards on human health. Front Genet 2024; 14:1306600. [PMID: 38299096 PMCID: PMC10829887 DOI: 10.3389/fgene.2023.1306600] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Accepted: 11/20/2023] [Indexed: 02/02/2024] Open
Abstract
Environmental pollution nowadays has not only a direct correlation with human health changes but a direct social impact. Epidemiological studies have evidenced the increased damage to human health on a daily basis because of damage to the ecological niche. Rapid urban growth and industrialized societies importantly compromise air quality, which can be assessed by a notable accumulation of air pollutants in both the gas and the particle phases. Of them, particulate matter (PM) represents a highly complex mixture of organic and inorganic compounds of the most variable size, composition, and origin. PM being one of the most complex environmental pollutants, its accumulation also varies in a temporal and spatial manner, which challenges current analytical techniques used to investigate PM interactions. Nevertheless, the characterization of the chemical composition of PM is a reliable indicator of the composition of the atmosphere, the quality of breathed air in urbanized societies, industrial zones and consequently gives support for pertinent measures to avoid serious health damage. Epigenomic damage is one of the most promising biological mechanisms of air pollution-derived carcinogenesis. Therefore, this review aims to highlight the implication of PM exposure in diverse molecular mechanisms driving human diseases by altered epigenetic regulation. The presented findings in the context of pan-organic cancer, fibrosis, neurodegeneration and metabolic diseases may provide valuable insights into the toxicity effects of PM components at the epigenomic level and may serve as biomarkers of early detection for novel targeted therapies.
Collapse
Affiliation(s)
- Dulcemaría Gavito-Covarrubias
- International Laboratory EPIGEN, Consejo de Ciencia y Tecnología del Estado de Puebla (CONCYTEP), Instituto de Ciencias, Benemérita Universidad Autónoma de Puebla (BUAP), Puebla, Puebla, Mexico
| | - Ivonne Ramírez-Díaz
- International Laboratory EPIGEN, Consejo de Ciencia y Tecnología del Estado de Puebla (CONCYTEP), Instituto de Ciencias, Benemérita Universidad Autónoma de Puebla (BUAP), Puebla, Puebla, Mexico
- Universidad Popular Autónoma del Estado de Puebla (UPAEP), Puebla, Mexico
| | - Josué Guzmán-Linares
- International Laboratory EPIGEN, Consejo de Ciencia y Tecnología del Estado de Puebla (CONCYTEP), Instituto de Ciencias, Benemérita Universidad Autónoma de Puebla (BUAP), Puebla, Puebla, Mexico
| | - Ilhuicamina Daniel Limón
- Laboratory of Neuropharmacology, Faculty of Chemical Sciences, Benemérita Universidad Autónoma de Puebla (BUAP), Puebla, Mexico
| | - Dulce María Manuel-Sánchez
- Laboratory of Neuropharmacology, Faculty of Chemical Sciences, Benemérita Universidad Autónoma de Puebla (BUAP), Puebla, Mexico
| | - Alejandro Molina-Herrera
- International Laboratory EPIGEN, Consejo de Ciencia y Tecnología del Estado de Puebla (CONCYTEP), Instituto de Ciencias, Benemérita Universidad Autónoma de Puebla (BUAP), Puebla, Puebla, Mexico
| | - Miguel Ángel Coral-García
- International Laboratory EPIGEN, Consejo de Ciencia y Tecnología del Estado de Puebla (CONCYTEP), Instituto de Ciencias, Benemérita Universidad Autónoma de Puebla (BUAP), Puebla, Puebla, Mexico
| | - Estela Anastasio
- International Laboratory EPIGEN, Consejo de Ciencia y Tecnología del Estado de Puebla (CONCYTEP), Instituto de Ciencias, Benemérita Universidad Autónoma de Puebla (BUAP), Puebla, Puebla, Mexico
| | - Arely Anaya-Hernández
- Centro de Investigación en Genética y Ambiente, Universidad Autónoma de Tlaxcala, Tlaxcala, Mexico
| | - Primavera López-Salazar
- Centro de Investigaciones en Dispositivos Semiconductores (CIDS), Instituto de Ciencias, Benemérita Universidad Autónoma de Puebla (BUAP), Puebla, Mexico
| | - Gabriel Juárez-Díaz
- Centro de Investigaciones en Dispositivos Semiconductores (CIDS), Instituto de Ciencias, Benemérita Universidad Autónoma de Puebla (BUAP), Puebla, Mexico
| | - Javier Martínez-Juárez
- Centro de Investigaciones en Dispositivos Semiconductores (CIDS), Instituto de Ciencias, Benemérita Universidad Autónoma de Puebla (BUAP), Puebla, Mexico
| | - Julián Torres-Jácome
- Laboratorio de Fisiopatología Cardiovascular, Instituto de Ciencias, Benemérita Universidad Autónoma de Puebla (BUAP), Puebla, Mexico
| | - Alondra Albarado-Ibáñez
- Laboratorio de Fisiopatología Cardiovascular, Instituto de Ciencias, Benemérita Universidad Autónoma de Puebla (BUAP), Puebla, Mexico
| | - Ygnacio Martínez-Laguna
- Vicerrectoría de Investigación y Estudios de Posgrado, Benemérita Universidad Autónoma de Puebla (BUAP), Puebla, Mexico
| | - Carolina Morán
- Centro de Investigación en Fisicoquímica de Materiales, Instituto de Ciencias, Benemérita Universidad Autónoma de Puebla (BUAP), Puebla, Mexico
| | - Karla Rubio
- International Laboratory EPIGEN, Consejo de Ciencia y Tecnología del Estado de Puebla (CONCYTEP), Instituto de Ciencias, Benemérita Universidad Autónoma de Puebla (BUAP), Puebla, Puebla, Mexico
| |
Collapse
|
76
|
Pérez-Martínez L, Romero L, Verdugo-Sivianes EM, Muñoz-Galván S, Rubio-Mediavilla S, Amiama-Roig A, Carnero A, Blanco JR. Role of maraviroc and/or rapamycin in the liver of IL10 KO mice with frailty syndrome. PLoS One 2024; 19:e0286201. [PMID: 38198476 PMCID: PMC10781157 DOI: 10.1371/journal.pone.0286201] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Accepted: 05/10/2023] [Indexed: 01/12/2024] Open
Abstract
Cellular senescence and low-grade inflammation favor the acceleration of aging. The liver is an essential metabolic organ because changes related to its function are related to age-related diseases. The objective of this study was to evaluate the effects of maraviroc (MVC) and/or rapamycin (RAPA) on liver tissue in an experimental model of frailty syndrome in mice, since MVC and RAPA are two molecules able to decrease CCR5 expression, which is overexpressed in patients with frailty. Methods: Eighty male homozygous IL10KO mice were randomly assigned to one of 4 groups (n = 20): i) IL10KO group; ii) MVC group, iii) RAPA group, and iv) MVC-RAPA group. Liver samples were analyzed. Gene expression quantification and western blotting were also performed. The proinflammatory cytokines IL-6 and IL-18 were decreased in MVC and MVC/RAPA groups, IL-12 was decreased in RAPA and MVC/RAPA groups and TNF-α was decreased in all therapeutic groups. P21 was decreased in RAPA and MVC/RAPA groups, Galactosidase beta-1, was also significantly reduced in all therapeutic groups, as were NF-kB1, NF-kB2 and STAT3. In all groups, mTOR and CCL5 were significantly reduced. CCR5 expression was decreased in the MVC and MVC/RAPA groups. Conclusion: MVC and RAPA may protect against some factors involved in liver aging. More studies will be necessary to verify their clinical applications.
Collapse
Affiliation(s)
| | - Lourdes Romero
- Centro de Investigación Biomédica de La Rioja (CIBIR), Logroño, Spain
| | - Eva M. Verdugo-Sivianes
- Instituto de Biomedicina de Sevilla, IBIS, Hospital Universitario Virgen del Rocío, Consejo Superior de Investigaciones Científicas, Universidad de Sevilla, Sevilla, Spain
- CIBERONC, Instituto de Salud Carlos III, Madrid, Spain
| | - Sandra Muñoz-Galván
- Instituto de Biomedicina de Sevilla, IBIS, Hospital Universitario Virgen del Rocío, Consejo Superior de Investigaciones Científicas, Universidad de Sevilla, Sevilla, Spain
- CIBERONC, Instituto de Salud Carlos III, Madrid, Spain
| | | | - Ana Amiama-Roig
- Centro de Investigación Biomédica de La Rioja (CIBIR), Logroño, Spain
| | - Amancio Carnero
- Instituto de Biomedicina de Sevilla, IBIS, Hospital Universitario Virgen del Rocío, Consejo Superior de Investigaciones Científicas, Universidad de Sevilla, Sevilla, Spain
- CIBERONC, Instituto de Salud Carlos III, Madrid, Spain
| | - José-Ramón Blanco
- Centro de Investigación Biomédica de La Rioja (CIBIR), Logroño, Spain
- Servicio de Enfermedades Infecciosas, Hospital Universitario San Pedro, Logroño, Spain
| |
Collapse
|
77
|
Lei Z, Lin W. Mechanisms Governing Oligodendrocyte Viability in Multiple Sclerosis and Its Animal Models. Cells 2024; 13:116. [PMID: 38247808 PMCID: PMC10814231 DOI: 10.3390/cells13020116] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Revised: 01/04/2024] [Accepted: 01/05/2024] [Indexed: 01/23/2024] Open
Abstract
Multiple sclerosis (MS) is a chronic autoimmune inflammatory demyelinating disease of the central nervous system (CNS), which is triggered by an autoimmune assault targeting oligodendrocytes and myelin. Recent research indicates that the demise of oligodendrocytes due to an autoimmune attack contributes significantly to the pathogenesis of MS and its animal model experimental autoimmune encephalomyelitis (EAE). A key challenge in MS research lies in comprehending the mechanisms governing oligodendrocyte viability and devising therapeutic approaches to enhance oligodendrocyte survival. Here, we provide an overview of recent findings that highlight the contributions of oligodendrocyte death to the development of MS and EAE and summarize the current literature on the mechanisms governing oligodendrocyte viability in these diseases.
Collapse
Affiliation(s)
- Zhixin Lei
- School of Chemistry, Chemical Engineering and Life Science, Wuhan University of Technology, Wuhan 430070, China;
| | - Wensheng Lin
- Department of Neuroscience, University of Minnesota, Minneapolis, MN 55455, USA
- Institute for Translational Neuroscience, University of Minnesota, Minneapolis, MN 55455, USA
| |
Collapse
|
78
|
Tuo H, Li W, Zhao W, Zhao J, Li D, Jin L. Shikonin alleviates doxorubicin-induced cardiotoxicity via Mst1/Nrf2 pathway in mice. Sci Rep 2024; 14:924. [PMID: 38195835 PMCID: PMC10776756 DOI: 10.1038/s41598-024-51675-7] [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: 09/03/2023] [Accepted: 01/08/2024] [Indexed: 01/11/2024] Open
Abstract
Doxorubicin (DOX) is a popular and potent anticancer drug, but its cardiotoxicity limits its clinical application. Shikonin has a wide range of biological functions, including antioxidant and anti-inflammatory effects. The aim of this study was to investigate the effects of shikonin on DOX-induced cardiac injury and to identify the underlying mechanisms. Mice receiving shikonin showed reduced cardiac injury response and enhanced cardiac function after DOX administration. Shikonin significantly attenuated DOX-induced oxidative damage, inflammation accumulation and cardiomyocyte apoptosis. Shikonin protects against DOX-induced cardiac injury by inhibiting Mammalian sterile 20-like kinase 1 (Mst1) and oxidative stress and activating the nuclear factor erythroid 2-related factor 2 (Nrf2) pathway. In conclusion, shikonin alleviates DOX-induced cardiotoxicity by inhibiting Mst1 and activating Nrf2. Shikonin may be used to treat DOX-induced cardiac injury.
Collapse
Affiliation(s)
- Hu Tuo
- Department of Pediatrics, Renmin Hospital of Wuhan University, Wuhan, China
| | - Wenjing Li
- Department of Pediatrics, Renmin Hospital of Wuhan University, Wuhan, China
| | - Wei Zhao
- Department of Pediatrics, Renmin Hospital of Wuhan University, Wuhan, China
| | - Juan Zhao
- Department of Pediatrics, Renmin Hospital of Wuhan University, Wuhan, China
| | - Danni Li
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Lin Jin
- Department of Orthopedics, Renmin Hospital of Wuhan University, Jiefang Road 238, Wuhan, 430060, China.
| |
Collapse
|
79
|
李 溢, 王 洪, 李 丹, 王 秋. [Distribution characteristics and correlation analysis of GJB2 variation in patients with auditory neuropathy]. LIN CHUANG ER BI YAN HOU TOU JING WAI KE ZA ZHI = JOURNAL OF CLINICAL OTORHINOLARYNGOLOGY, HEAD, AND NECK SURGERY 2024; 38:23-29. [PMID: 38297845 PMCID: PMC11116162 DOI: 10.13201/j.issn.2096-7993.2024.01.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 10/15/2023] [Indexed: 02/02/2024]
Abstract
Objective:To elucidate the correlation between the GJB2 gene and auditory neuropathy, aiming to provide valuable insights for genetic counseling of affected individuals and their families. Methods:The general information, audiological data(including pure tone audiometry, distorted otoacoustic emission, auditory brainstem response, electrocochlography), imaging data and genetic test data of 117 auditory neuropathy patients, and the patients with GJB2 gene mutation were screened out for the correlation analysis of auditory neuropathy. Results:Total of 16 patients were found to have GJB2 gene mutations, all of which were pathogenic or likely pathogenic.was Among them, one patient had compound heterozygous variants GJB2[c. 427C>T][c. 358_360del], exhibiting total deafness. One was GJB2[c. 299_300delAT][c. 35_36insG]compound heterozygous variants, the audiological findings were severe hearing loss.The remaining 14 patients with GJB2 gene variants exhibited typical auditory neuropathy. Conclusion:In this study, the relationship between GJB2 gene and auditory neuropathy was preliminarily analyzed,and explained the possible pathogenic mechanism of GJB2 gene variants that may be related to auditory neuropathy.
Collapse
Affiliation(s)
- 溢铭 李
- 中国人民解放军总医院第六医学中心耳鼻咽喉头颈外科医学部耳鼻咽喉内科解放军医学院(北京,100048)Department of Audiology and Vestibular Medicine, College of Otolaryngology Head and Neck Surgery Department of Otolaryngology, the Sixth Medical Centre, Chinese PLA General Hospital, Medical School of Chinese PLA, Beijing, 100048, China
- 国家耳鼻咽喉疾病临床医学研究中心National Clinical Medical Center for Otolaryngology
| | - 洪阳 王
- 中国人民解放军总医院第六医学中心耳鼻咽喉头颈外科医学部耳鼻咽喉内科解放军医学院(北京,100048)Department of Audiology and Vestibular Medicine, College of Otolaryngology Head and Neck Surgery Department of Otolaryngology, the Sixth Medical Centre, Chinese PLA General Hospital, Medical School of Chinese PLA, Beijing, 100048, China
- 国家耳鼻咽喉疾病临床医学研究中心National Clinical Medical Center for Otolaryngology
| | - 丹阳 李
- 中国人民解放军总医院第六医学中心耳鼻咽喉头颈外科医学部耳鼻咽喉内科解放军医学院(北京,100048)Department of Audiology and Vestibular Medicine, College of Otolaryngology Head and Neck Surgery Department of Otolaryngology, the Sixth Medical Centre, Chinese PLA General Hospital, Medical School of Chinese PLA, Beijing, 100048, China
- 国家耳鼻咽喉疾病临床医学研究中心National Clinical Medical Center for Otolaryngology
| | - 秋菊 王
- 中国人民解放军总医院第六医学中心耳鼻咽喉头颈外科医学部耳鼻咽喉内科解放军医学院(北京,100048)Department of Audiology and Vestibular Medicine, College of Otolaryngology Head and Neck Surgery Department of Otolaryngology, the Sixth Medical Centre, Chinese PLA General Hospital, Medical School of Chinese PLA, Beijing, 100048, China
- 国家耳鼻咽喉疾病临床医学研究中心National Clinical Medical Center for Otolaryngology
| |
Collapse
|
80
|
Kim Y, Lee SB, Cho M, Choe S, Jang M. Indian Almond ( Terminalia catappa Linn.) Leaf Extract Extends Lifespan by Improving Lipid Metabolism and Antioxidant Activity Dependent on AMPK Signaling Pathway in Caenorhabditis elegans under High-Glucose-Diet Conditions. Antioxidants (Basel) 2023; 13:14. [PMID: 38275634 PMCID: PMC10812731 DOI: 10.3390/antiox13010014] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Revised: 12/13/2023] [Accepted: 12/14/2023] [Indexed: 01/27/2024] Open
Abstract
This study aimed to evaluate the antioxidant and antiaging effects of Indian almond (Terminalia catappa Linn.) leaf extract (TCE) on high-glucose (GLU)-induced obese Caenorhabditis elegans. Since TCE contains high contents of flavonoids and phenolics, strong radical scavenging activity was confirmed in vitro. The stress-resistance effect of TCE was confirmed under thermal and oxidative stress conditions at nontoxic tested concentrations (6.25, 12.5, and 25 μg/mL). GLU at 2% caused lipid and reactive oxygen species (ROS) accumulation in C. elegans, and TCE inhibited lipid and ROS accumulation under both normal and 2% GLU conditions in a concentration-dependent manner. In addition, TCE proved to be effective in prolonging the lifespan of C. elegans under normal and 2% GLU conditions. The ROS reduction effect of TCE was abolished in mutants deficient in daf-16/FOXO and skn-1/Nrf-2. In addition, the lifespan-extending effect of TCE in these two mutants disappeared. The lifespan-extending effect was abolished even in atgl-1/ATGL-deficiency mutants. The TCE effect was reduced in aak-1/AMPK-deficient mutants and completely abolished under 2% GLU conditions. Therefore, the effect of prolonging lifespan by inhibiting lipid and ROS accumulation under the high GLU conditions of TCE is considered to be the result of atgl-1, daf-16, and skn-1 being downregulated by aak-1. These results suggest that the physiological potential of TCE contributes to antiaging under metabolic disorders.
Collapse
Affiliation(s)
- Yebin Kim
- Department of Smart Food and Drug, Inje University, Gimhae 50834, Republic of Korea; (Y.K.); (S.-b.L.); (M.C.)
| | - Seul-bi Lee
- Department of Smart Food and Drug, Inje University, Gimhae 50834, Republic of Korea; (Y.K.); (S.-b.L.); (M.C.)
| | - Myogyeong Cho
- Department of Smart Food and Drug, Inje University, Gimhae 50834, Republic of Korea; (Y.K.); (S.-b.L.); (M.C.)
| | - Soojin Choe
- Department of Food Technology and Nutrition, Inje University, Gimhae 50834, Republic of Korea;
| | - Miran Jang
- Department of Smart Food and Drug, Inje University, Gimhae 50834, Republic of Korea; (Y.K.); (S.-b.L.); (M.C.)
- Department of Food Technology and Nutrition, Inje University, Gimhae 50834, Republic of Korea;
| |
Collapse
|
81
|
Wang W, Zhang J. Teneligliptin alleviates diabetes-related cognitive impairment by inhibiting the endoplasmic reticulum (ER) stress and NLRP3 inflammasome in mice. Aging (Albany NY) 2023; 16:8336-8347. [PMID: 38127000 PMCID: PMC11131981 DOI: 10.18632/aging.205333] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Accepted: 10/23/2023] [Indexed: 12/23/2023]
Abstract
Diabetes mellitus (DM) significantly influences the normal health of patients with its severe complications, including diabetes-related cognitive impairment (CI). Recently, neuroinflammation and oxidative stress (OS) have been reported to participate in the pathogenesis of diabetes-related CI. Teneligliptin, an inhibitor of DDP-IV, was developed for treating DM and is claimed with promising effects against inflammation. Herein, in the current study, we examined the potential therapeutic function of Teneligliptin against diabetes-related CI. Db/m or diabetic mice were orally administered with teneligliptin (60 mg/kg/day) for 10 weeks. Elevated levels of total cholesterol (TC), triglyceride (TG), and low-density lipoprotein cholesterol (LDL-C), increased escape latency, declined time in the platform quadrant and decreased number of platform crossings in the Morris water maze test, reduced freezing index in the fear conditioning test, and lessened time spent in the novel arm and percentage of alterations in the Y-maze test were observed in diabetic mice, all of which were sharply improved by teneligliptin. Furthermore, increased levels of inflammatory cytokines and activated OS state were observed in the hippocampus of diabetic mice, which were markedly repressed by Teneligliptin. Lastly, the activation of the NOD-like receptor family pyrin domain containing 3 (NLRP3) signaling and the endoplasmic reticulum (ER) stress pathway in the hippocampus of diabetic mice were notably inhibited by teneligliptin. Collectively, teneligliptin mitigated diabetes-related CI by repressing the ER stress and NLRP3 inflammasome in diabetic mice.
Collapse
Affiliation(s)
- Weifeng Wang
- Department of Endocrinology, Laizhou City People’s Hospital, Yantai, Shandong 261400, China
| | - Juanjuan Zhang
- Department of Endocrinology, Laizhou City People’s Hospital, Yantai, Shandong 261400, China
| |
Collapse
|
82
|
Chen Q, Liu R, Wei C, Wang X, Wu X, Fan R, Yu X, Li Z, Mao R, Hu J, Zhu N, Liu X, Li Y, Xu M. Exogenous Nucleotides Ameliorate Age-Related Decline in Testosterone in Male Senescence-Accelerated Mouse Prone-8 (SAMP8) Mice by Modulating the Local Renin-Angiotensin System Antioxidant Pathway. Nutrients 2023; 15:5130. [PMID: 38140389 PMCID: PMC10745527 DOI: 10.3390/nu15245130] [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: 10/31/2023] [Revised: 12/02/2023] [Accepted: 12/12/2023] [Indexed: 12/24/2023] Open
Abstract
In older men, an age-related decline in testosterone is closely associated with various adverse health outcomes. With the progression of aging, hyperactivation of the local renin-angiotensin system (RAS) and oxidative stress increase in the testis. The regulation of RAS antioxidants may be a target to delay testicular aging and maintain testosterone levels. Exogenous nucleotides (NTs) have anti-aging potential in several systems, but there are no studies of their effects on the reproductive system. In our study, we examined the effects of exogenous NTs on testosterone synthesis and explored possible mechanisms of action. Therefore, senescence-accelerated mouse prone-8 (SAMP8) mice and senescence-accelerated mouse resistant 1 (SAMR1) were used in the experiment, and they were randomly divided into an NTs free group (NTs-F), a normal control group (control), a low-dose NTs group (NTs-L), a middle-dose NTs (NTs-M), a high-dose NTs group (NTs-H) and SAMR1 groups, and the testis of the mice were collected for testing after 9 months of intervention. The results showed that exogenous NTs could increase the testicular organ index in mice during aging, and delayed the age-associated decline in testosterone levels in SAMP8 male mice, possibly by modulating the local RAS antioxidant pathway and reducing oxidative stress to protect the testis. The present study provides new research clues for the development of preventive and therapeutic strategies for related diseases.
Collapse
Affiliation(s)
- Qianqian Chen
- Department of Nutrition and Food Hygiene, School of Public Health, Peking University, Beijing 100191, China
- Beijing Key Laboratory of Toxicological Research and Risk Assessment for Food Safety, Peking University, Beijing 100191, China
| | - Rui Liu
- Department of Nutrition and Food Hygiene, School of Public Health, Peking University, Beijing 100191, China
- Beijing Key Laboratory of Toxicological Research and Risk Assessment for Food Safety, Peking University, Beijing 100191, China
| | - Chan Wei
- Department of Nutrition and Food Hygiene, School of Public Health, Peking University, Beijing 100191, China
- Beijing Key Laboratory of Toxicological Research and Risk Assessment for Food Safety, Peking University, Beijing 100191, China
| | - Xiujuan Wang
- Department of Nutrition and Food Hygiene, School of Public Health, Peking University, Beijing 100191, China
- Beijing Key Laboratory of Toxicological Research and Risk Assessment for Food Safety, Peking University, Beijing 100191, China
| | - Xin Wu
- Department of Nutrition and Food Hygiene, School of Public Health, Peking University, Beijing 100191, China
- Beijing Key Laboratory of Toxicological Research and Risk Assessment for Food Safety, Peking University, Beijing 100191, China
| | - Rui Fan
- Department of Nutrition and Food Hygiene, School of Public Health, Peking University, Beijing 100191, China
- Beijing Key Laboratory of Toxicological Research and Risk Assessment for Food Safety, Peking University, Beijing 100191, China
| | - Xiaochen Yu
- Department of Nutrition and Food Hygiene, School of Public Health, Peking University, Beijing 100191, China
- Beijing Key Laboratory of Toxicological Research and Risk Assessment for Food Safety, Peking University, Beijing 100191, China
| | - Zhen Li
- Department of Nutrition and Food Hygiene, School of Public Health, Peking University, Beijing 100191, China
- Beijing Key Laboratory of Toxicological Research and Risk Assessment for Food Safety, Peking University, Beijing 100191, China
| | - Ruixue Mao
- Department of Nutrition and Food Hygiene, School of Public Health, Peking University, Beijing 100191, China
- Beijing Key Laboratory of Toxicological Research and Risk Assessment for Food Safety, Peking University, Beijing 100191, China
| | - Jiani Hu
- Department of Nutrition and Food Hygiene, School of Public Health, Peking University, Beijing 100191, China
- Beijing Key Laboratory of Toxicological Research and Risk Assessment for Food Safety, Peking University, Beijing 100191, China
| | - Na Zhu
- Department of Nutrition and Food Hygiene, School of Public Health, Peking University, Beijing 100191, China
- Beijing Key Laboratory of Toxicological Research and Risk Assessment for Food Safety, Peking University, Beijing 100191, China
| | - Xinran Liu
- Department of Nutrition and Food Hygiene, School of Public Health, Peking University, Beijing 100191, China
- Beijing Key Laboratory of Toxicological Research and Risk Assessment for Food Safety, Peking University, Beijing 100191, China
| | - Yong Li
- Department of Nutrition and Food Hygiene, School of Public Health, Peking University, Beijing 100191, China
- Beijing Key Laboratory of Toxicological Research and Risk Assessment for Food Safety, Peking University, Beijing 100191, China
| | - Meihong Xu
- Department of Nutrition and Food Hygiene, School of Public Health, Peking University, Beijing 100191, China
- Beijing Key Laboratory of Toxicological Research and Risk Assessment for Food Safety, Peking University, Beijing 100191, China
| |
Collapse
|
83
|
Yan Y, Yu L, Chen B, Cao C, Zhao H, Wang Q, Xie D, Xi Y, Zhang C, Cheng J. Mastoparan M Suppressed NLRP3 Inflammasome Activation by Inhibiting MAPK/NF-κB and Oxidative Stress in Gouty Arthritis. J Inflamm Res 2023; 16:6179-6193. [PMID: 38116368 PMCID: PMC10730329 DOI: 10.2147/jir.s434587] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Accepted: 11/28/2023] [Indexed: 12/21/2023] Open
Abstract
Background Gouty arthritis is characterized by the accumulation of monosodium urate crystals (MSU) in the synovial joints and surrounding tissues. Mastoparan M (Mast-M) is a biologically active peptide composed of 14 amino acids, extracted from wasp venom. This study aims to assess the impact of Mast-M on in vitro and in vivo gouty arthritis induced by lipolyaccharide (LPS) plus MSU crystal stimulation. Methods PMA-differentiated THP-1 macrophages were pre-treated with Mast-M or left untreated, followed by stimulation with LPS and MSU crystals. Cell lysates were collected to assess the expression of the NLRP3 inflammasome, inflammatory signaling pathways, and oxidative stress. Furthermore, to evaluate the in vivo anti-inflammatory effect of Mast-M, an experimental acute gouty arthritis mouse model was established through intra-articular injection of MSU crystals. Results Mast-M treatment demonstrated significant inhibition of the phosphorylation of MAPKs/NF-κB signaling pathways and reduction in oxidative stress expression in LPS and MSU-induced THP-1 macrophages. This resulted in the suppression of downstream NLRP3 inflammasome activation and IL-1β release. In vivo, Mast-M effectively attenuated the inflammation induced by MSU in mice with gouty arthritis. Specifically, Mast-M reduced swelling in the paws, inhibited the infiltration of neutrophils and macrophages into periarticular tissue, and decreased the activation of the NLRP3 inflammasome and IL-1β production. Conclusion Mast-M significantly improves gouty arthritis, and its potential mechanism may be achieved by inhibiting the MAPK/NF-κB pathway and alleviating oxidative stress, thus suppressing the activation of NLRP3 inflammasomes.
Collapse
Affiliation(s)
- Yunbo Yan
- Department of Internal Medicine, Xiang’an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, People’s Republic of China
| | - Linqian Yu
- Department of Internal Medicine, Xiang’an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, People’s Republic of China
| | - Binyang Chen
- Department of Internal Medicine, Xiang’an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, People’s Republic of China
| | - Chang’an Cao
- Department of Internal Medicine, Xiang’an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, People’s Republic of China
| | - Hairong Zhao
- Department of Internal Medicine, Xiang’an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, People’s Republic of China
- Yunnan Provincial Key Laboratory of Entomological Biopharmaceutical R&D, Dali University, Dali, People’s Republic of China
| | - Qiang Wang
- Department of Internal Medicine, Xiang’an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, People’s Republic of China
| | - De Xie
- Department of Internal Medicine, Xiang’an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, People’s Republic of China
| | - Yuemei Xi
- Department of Internal Medicine, Xiang’an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, People’s Republic of China
| | - Chenggui Zhang
- Yunnan Provincial Key Laboratory of Entomological Biopharmaceutical R&D, Dali University, Dali, People’s Republic of China
| | - Jidong Cheng
- Department of Internal Medicine, Xiang’an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, People’s Republic of China
- Xiamen Key Laboratory of Translational Medicine for Nucleic Acid Metabolism and Regulation, Xiamen, People’s Republic of China
| |
Collapse
|
84
|
Thomas C, Erni R, Wu JY, Fischer F, Lamers G, Grigolon G, Mitchell SJ, Zarse K, Carreira EM, Ristow M. A naturally occurring polyacetylene isolated from carrots promotes health and delays signatures of aging. Nat Commun 2023; 14:8142. [PMID: 38065964 PMCID: PMC10709416 DOI: 10.1038/s41467-023-43672-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Accepted: 11/16/2023] [Indexed: 12/18/2023] Open
Abstract
To ameliorate or even prevent signatures of aging in ultimately humans, we here report the identification of a previously undescribed polyacetylene contained in the root of carrots (Daucus carota), hereafter named isofalcarintriol, which we reveal as potent promoter of longevity in the nematode C. elegans. We assign the absolute configuration of the compound as (3 S,8 R,9 R,E)-heptadeca-10-en-4,6-diyne-3,8,9-triol, and develop a modular asymmetric synthesis route for all E-isofalcarintriol stereoisomers. At the molecular level, isofalcarintriol affects cellular respiration in mammalian cells, C. elegans, and mice, and interacts with the α-subunit of the mitochondrial ATP synthase to promote mitochondrial biogenesis. Phenotypically, this also results in decreased mammalian cancer cell growth, as well as improved motility and stress resistance in C. elegans, paralleled by reduced protein accumulation in nematodal models of neurodegeneration. In addition, isofalcarintriol supplementation to both wild-type C57BL/6NRj mice on high-fat diet, and aged mice on chow diet results in improved glucose metabolism, increased exercise endurance, and attenuated parameters of frailty at an advanced age. Given these diverse effects on health parameters in both nematodes and mice, isofalcarintriol might become a promising mitohormesis-inducing compound to delay, ameliorate, or prevent aging-associated diseases in humans.
Collapse
Affiliation(s)
- Carolin Thomas
- Laboratory of Energy Metabolism, Institute of Translational Medicine, Department of Health Sciences and Technology, Swiss Federal Institute (ETH) Zurich, Schorenstrasse 16, 8603, Schwerzenbach, Switzerland
| | - Reto Erni
- Laboratory of Chemistry and Applied Biosciences, Department of Organic Chemistry, Swiss Federal Institute (ETH) Zurich, Vladimir-Prelog-Weg 1-5/10, Zurich, 8093, Switzerland
- Biozentrum, University of Basel, Basel, 4056, Switzerland
| | - Jia Yee Wu
- Laboratory of Energy Metabolism, Institute of Translational Medicine, Department of Health Sciences and Technology, Swiss Federal Institute (ETH) Zurich, Schorenstrasse 16, 8603, Schwerzenbach, Switzerland
| | - Fabian Fischer
- Laboratory of Energy Metabolism, Institute of Translational Medicine, Department of Health Sciences and Technology, Swiss Federal Institute (ETH) Zurich, Schorenstrasse 16, 8603, Schwerzenbach, Switzerland
- CureVac SE, Tübingen, 72076, Germany
| | - Greta Lamers
- Laboratory of Energy Metabolism, Institute of Translational Medicine, Department of Health Sciences and Technology, Swiss Federal Institute (ETH) Zurich, Schorenstrasse 16, 8603, Schwerzenbach, Switzerland
| | - Giovanna Grigolon
- Laboratory of Energy Metabolism, Institute of Translational Medicine, Department of Health Sciences and Technology, Swiss Federal Institute (ETH) Zurich, Schorenstrasse 16, 8603, Schwerzenbach, Switzerland
| | - Sarah J Mitchell
- Ludwig Princeton Branch, Princeton University, Princeton, NJ, 08540, USA
| | - Kim Zarse
- Laboratory of Energy Metabolism, Institute of Translational Medicine, Department of Health Sciences and Technology, Swiss Federal Institute (ETH) Zurich, Schorenstrasse 16, 8603, Schwerzenbach, Switzerland
- Institute of Experimental Endocrinology, Charité Universitätsmedizin Berlin, Berlin, 10117, Germany
| | - Erick M Carreira
- Laboratory of Chemistry and Applied Biosciences, Department of Organic Chemistry, Swiss Federal Institute (ETH) Zurich, Vladimir-Prelog-Weg 1-5/10, Zurich, 8093, Switzerland.
| | - Michael Ristow
- Laboratory of Energy Metabolism, Institute of Translational Medicine, Department of Health Sciences and Technology, Swiss Federal Institute (ETH) Zurich, Schorenstrasse 16, 8603, Schwerzenbach, Switzerland.
- Institute of Experimental Endocrinology, Charité Universitätsmedizin Berlin, Berlin, 10117, Germany.
| |
Collapse
|
85
|
Liang J, Huang F, Song Z, Tang R, Zhang P, Chen R. Impact of NAD+ metabolism on ovarian aging. Immun Ageing 2023; 20:70. [PMID: 38041117 PMCID: PMC10693113 DOI: 10.1186/s12979-023-00398-w] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Accepted: 11/21/2023] [Indexed: 12/03/2023]
Abstract
Nicotinamide adenine dinucleotide (NAD+), a crucial coenzyme in cellular redox reactions, is closely associated with age-related functional degeneration and metabolic diseases. NAD exerts direct and indirect influences on many crucial cellular functions, including metabolic pathways, DNA repair, chromatin remodeling, cellular senescence, and immune cell functionality. These cellular processes and functions are essential for maintaining tissue and metabolic homeostasis, as well as healthy aging. Causality has been elucidated between a decline in NAD levels and multiple age-related diseases, which has been confirmed by various strategies aimed at increasing NAD levels in the preclinical setting. Ovarian aging is recognized as a natural process characterized by a decline in follicle number and function, resulting in decreased estrogen production and menopause. In this regard, it is necessary to address the many factors involved in this complicated procedure, which could improve fertility in women of advanced maternal age. Concerning the decrease in NAD+ levels as ovarian aging progresses, promising and exciting results are presented for strategies using NAD+ precursors to promote NAD+ biosynthesis, which could substantially improve oocyte quality and alleviate ovarian aging. Hence, to acquire further insights into NAD+ metabolism and biology, this review aims to probe the factors affecting ovarian aging, the characteristics of NAD+ precursors, and the current research status of NAD+ supplementation in ovarian aging. Specifically, by gaining a comprehensive understanding of these aspects, we are optimistic about the prominent progress that will be made in both research and therapy related to ovarian aging.
Collapse
Affiliation(s)
- Jinghui Liang
- Department of Obstetrics and Gynecology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, National Clinical Research Center for Obstetric & Gynecologic Diseases, Beijing, 100730, China
| | - Feiling Huang
- Department of Obstetrics and Gynecology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, National Clinical Research Center for Obstetric & Gynecologic Diseases, Beijing, 100730, China
| | - Zhaoqi Song
- School of Medical Technology and Engineering, Fujian Medical University, Fuzhou, Fujian, China
| | - Ruiyi Tang
- Department of Obstetrics and Gynecology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, National Clinical Research Center for Obstetric & Gynecologic Diseases, Beijing, 100730, China
| | - Peng Zhang
- Beijing Key Laboratory for Genetics of Birth Defects, Beijing Pediatric Research Institute, MOE Key Laboratory of Major Diseases in Children, Rare Disease Center, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, 100045, China.
| | - Rong Chen
- Department of Obstetrics and Gynecology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, National Clinical Research Center for Obstetric & Gynecologic Diseases, Beijing, 100730, China.
| |
Collapse
|
86
|
Lapenna D. Glutathione and glutathione-dependent enzymes: From biochemistry to gerontology and successful aging. Ageing Res Rev 2023; 92:102066. [PMID: 37683986 DOI: 10.1016/j.arr.2023.102066] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2023] [Revised: 08/24/2023] [Accepted: 09/04/2023] [Indexed: 09/10/2023]
Abstract
The tripeptide glutathione (GSH), namely γ-L-glutamyl-L-cysteinyl-glycine, is an ubiquitous low-molecular weight thiol nucleophile and reductant of utmost importance, representing the central redox agent of most aerobic organisms. GSH has vital functions involving also antioxidant protection, detoxification, redox homeostasis, cell signaling, iron metabolism/homeostasis, DNA synthesis, gene expression, cysteine/protein metabolism, and cell proliferation/differentiation or death including apoptosis and ferroptosis. Various functions of GSH are exerted in concert with GSH-dependent enzymes. Indeed, although GSH has direct scavenging antioxidant effects, its antioxidant function is substantially accomplished by glutathione peroxidase-catalyzed reactions with reductive removal of H2O2, organic peroxides such as lipid hydroperoxides, and peroxynitrite; to this antioxidant activity also contribute peroxiredoxins, enzymes further involved in redox signaling and chaperone activity. Moreover, the detoxifying function of GSH is basically exerted in conjunction with glutathione transferases, which have also antioxidant properties. GSH is synthesized in the cytosol by the ATP-dependent enzymes glutamate cysteine ligase (GCL), which catalyzes ligation of cysteine and glutamate forming γ-glutamylcysteine (γ-GC), and glutathione synthase, which adds glycine to γ-GC resulting in GSH formation; GCL is rate-limiting for GSH synthesis, as is the precursor amino acid cysteine, which may be supplemented as N-acetylcysteine (NAC), a therapeutically available compound. After its cell export, GSH is degraded extracellularly by the membrane-anchored ectoenzyme γ-glutamyl transferase, a process occurring, as GSH synthesis and export, in the γ-glutamyl cycle. GSH degradation occurs also intracellularly by the cytoplasmic enzymatic ChaC family of γ-glutamyl cyclotransferase. Synthesis and degradation of GSH, together with its export, translocation to cell organelles, utilization for multiple essential functions, and regeneration from glutathione disulfide by glutathione reductase, are relevant to GSH homeostasis and metabolism. Notably, GSH levels decline during aging, an alteration generally related to impaired GSH biosynthesis and leading to cell dysfunction. However, there is evidence of enhanced GSH levels in elderly subjects with excellent physical and mental health status, suggesting that heightened GSH may be a marker and even a causative factor of increased healthspan and lifespan. Such aspects, and much more including GSH-boosting substances administrable to humans, are considered in this state-of-the-art review, which deals with GSH and GSH-dependent enzymes from biochemistry to gerontology, focusing attention also on lifespan/healthspan extension and successful aging; the significance of GSH levels in aging is considered also in relation to therapeutic possibilities and supplementation strategies, based on the use of various compounds including NAC-glycine, aimed at increasing GSH and related defenses to improve health status and counteract aging processes in humans.
Collapse
Affiliation(s)
- Domenico Lapenna
- Dipartimento di Medicina e Scienze dell'Invecchiamento, and Laboratorio di Fisiopatologia dello Stress Ossidativo, Center for Advanced Studies and Technology (CAST, former CeSI-MeT, Center of Excellence on Aging), Università degli Studi "G. d'Annunzio" Chieti Pescara, U.O.C. Medicina Generale 2, Ospedale Clinicizzato "Santissima Annunziata", Via dei Vestini, 66100 Chieti, Italy.
| |
Collapse
|
87
|
Hassanein EHM, Ibrahim IM, Abd El-Maksoud MS, Abd El-Aziz MK, Abd-Alhameed EK, Althagafy HS. Targeting necroptosis in fibrosis. Mol Biol Rep 2023; 50:10471-10484. [PMID: 37910384 PMCID: PMC10676318 DOI: 10.1007/s11033-023-08857-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Accepted: 09/27/2023] [Indexed: 11/03/2023]
Abstract
Necroptosis, a type of programmed cell death that resembles necrosis, is now known to depend on a different molecular mechanism from apoptosis, according to several recent studies. Many efforts have reported the possible influence of necroptosis in human disorders and concluded the crucial role in the pathophysiology of various diseases, including liver diseases, renal injuries, cancers, and others. Fibrosis is the most common end-stage pathological cascade of several chronic inflammatory disorders. In this review, we explain the impact of necroptosis and fibrosis, for which necroptosis has been demonstrated to be a contributing factor. We also go over the inhibitors of necroptosis and how they have been applied to fibrosis models. This review helps to clarify the role of necroptosis in fibrosis and will encourage clinical efforts to target this pathway of programmed cell death.
Collapse
Affiliation(s)
- Emad H M Hassanein
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Al-Azhar University, Assiut, Egypt.
| | - Islam M Ibrahim
- Graduated Student, Faculty of Pharmacy, Al-Azhar University, Assiut Branch, Assiut, 71524, Egypt
| | - Mostafa S Abd El-Maksoud
- Graduated Student, Faculty of Pharmacy, Al-Azhar University, Assiut Branch, Assiut, 71524, Egypt
| | - Mostafa K Abd El-Aziz
- Graduated Student, Faculty of Pharmacy, Al-Azhar University, Assiut Branch, Assiut, 71524, Egypt
| | - Esraa K Abd-Alhameed
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Beni-Suef University, Beni-Suef, Egypt
| | - Hanan S Althagafy
- Department of Biochemistry, Faculty of Science, University of Jeddah, Jeddah, Saudi Arabia
| |
Collapse
|
88
|
Schlosser A, Helfenrath K, Wisniewsky M, Hinrichs K, Burmester T, Fabrizius A. The knockout of cytoglobin 1 in zebrafish (Danio rerio) alters lipid metabolism, iron homeostasis and oxidative stress response. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2023; 1870:119558. [PMID: 37549740 DOI: 10.1016/j.bbamcr.2023.119558] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Revised: 07/19/2023] [Accepted: 08/01/2023] [Indexed: 08/09/2023]
Abstract
Cytoglobin (Cygb) is an evolutionary ancient heme protein with yet unclear physiological function(s). Mammalian Cygb is ubiquitously expressed in all tissues and is proposed to be involved in reactive oxygen species (ROS) detoxification, nitric oxide (NO) metabolism and lipid-based signaling processes. Loss-of-function studies in mouse associate Cygb with apoptosis, inflammation, fibrosis, cardiovascular dysfunction or oncogenesis. In zebrafish (Danio rerio), two cygb genes exist, cytoglobin 1 (cygb1) and cytoglobin 2 (cygb2). Both have different coordination states and distinct expression sites within zebrafish tissues. The biological roles of the cygb paralogs are largely uncharacterized. We used a CRISPR/Cas9 genome editing approach and generated a knockout of the penta-coordinated cygb1 for in vivo analysis. Adult male cygb1 knockouts develop phenotypic abnormalities, including weight loss. To identify the molecular mechanisms underlying the occurrence of these phenotypes and differentiate between function and effect of the knockout we compared the transcriptomes of cygb1 knockout at different ages to age-matched wild-type zebrafish. We found that immune regulatory and cell cycle regulatory transcripts (e.g. tp53) were up-regulated in the cygb1 knockout liver. Additionally, the expression of transcripts involved in lipid metabolism and transport, the antioxidative defense and iron homeostasis was affected in the cygb1 knockout. Cygb1 may function as an anti-inflammatory and cytoprotective factor in zebrafish liver, and may be involved in lipid-, iron-, and ROS-dependent signaling.
Collapse
Affiliation(s)
- Annette Schlosser
- Institute of Cell and Systems Biology of Animals, University of Hamburg, D-20146 Hamburg, Germany
| | - Kathrin Helfenrath
- Institute of Cell and Systems Biology of Animals, University of Hamburg, D-20146 Hamburg, Germany
| | - Michelle Wisniewsky
- Institute of Cell and Systems Biology of Animals, University of Hamburg, D-20146 Hamburg, Germany
| | - Kira Hinrichs
- Institute of Cell and Systems Biology of Animals, University of Hamburg, D-20146 Hamburg, Germany
| | - Thorsten Burmester
- Institute of Cell and Systems Biology of Animals, University of Hamburg, D-20146 Hamburg, Germany
| | - Andrej Fabrizius
- Institute of Cell and Systems Biology of Animals, University of Hamburg, D-20146 Hamburg, Germany.
| |
Collapse
|
89
|
Singh A, Tiwari S, Singh S. Pirh2 modulates amyloid-β aggregation through the regulation of glucose-regulated protein 78 and chaperone-mediated signaling. J Cell Physiol 2023; 238:2841-2854. [PMID: 37882235 DOI: 10.1002/jcp.31134] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Revised: 09/14/2023] [Accepted: 09/21/2023] [Indexed: 10/27/2023]
Abstract
Amyloid-β (Aβ) protein aggregation in the brain is a pathological hallmark of Alzheimer's disease (AD) however, the underlying molecular mechanisms regulating amyloid aggregation are not well understood. Here, we studied the propitious role of E3 ubiquitin ligase Pirh2 in Aβ protein aggregation in view of its regulatory ligase activity in the ubiquitin-proteasome system employing both cellular and sporadic rodent models of AD. Pirh2 protein abundance was significantly increased during Streptozotocin (STZ) induced AD conditions, and transient silencing of Pirh2 significantly inhibited the Aβ aggregation and modified the dendrite morphology along with the substantial decrease in choline level in the differentiated neurons. MALDI-TOF/TOF, coimmunoprecipitation, and UbcH7-linked in vitro ubiquitylation analysis confirmed the high interaction of Pirh2 with chaperone GRP78. Furthermore, Pirh2 silencing inhibits the STZ induced altered level of endoplasmic reticulum stress and intracellular Ca2+ levels in neuronal N2a cells. Pirh2 silencing also inhibited the AD conditions related to the altered protein abundance of HSP90 and its co-chaperones which may collectively involve in the reduced burden of amyloid aggregates in neuronal cells. Pirh2 silencing further stabilized the nuclear translocation of phospho-Nrf2 and inhibited the altered level of autophagy factors. Taken together, our data indicated that Pirh2 is critically involved in STZ induced AD pathogenesis through its interaction with ER-chaperone GRP78, improves the neuronal connectivity, affects the altered level of chaperones, co-chaperones, & autophagic markers, and collectively inhibits the Aβ aggregation.
Collapse
Affiliation(s)
- Abhishek Singh
- Division of Toxicology and Experimental Medicine, CSIR-Central Drug Research Institute, Lucknow, India
- Academy of Scientific & Innovative Research (AcSIR), Ghaziabad, India
| | - Shubhangini Tiwari
- Division of Toxicology and Experimental Medicine, CSIR-Central Drug Research Institute, Lucknow, India
| | - Sarika Singh
- Division of Toxicology and Experimental Medicine, CSIR-Central Drug Research Institute, Lucknow, India
- Academy of Scientific & Innovative Research (AcSIR), Ghaziabad, India
| |
Collapse
|
90
|
Böhm EW, Buonfiglio F, Voigt AM, Bachmann P, Safi T, Pfeiffer N, Gericke A. Oxidative stress in the eye and its role in the pathophysiology of ocular diseases. Redox Biol 2023; 68:102967. [PMID: 38006824 PMCID: PMC10701459 DOI: 10.1016/j.redox.2023.102967] [Citation(s) in RCA: 65] [Impact Index Per Article: 32.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Revised: 11/10/2023] [Accepted: 11/14/2023] [Indexed: 11/27/2023] Open
Abstract
Oxidative stress occurs through an imbalance between the generation of reactive oxygen species (ROS) and the antioxidant defense mechanisms of cells. The eye is particularly exposed to oxidative stress because of its permanent exposure to light and due to several structures having high metabolic activities. The anterior part of the eye is highly exposed to ultraviolet (UV) radiation and possesses a complex antioxidant defense system to protect the retina from UV radiation. The posterior part of the eye exhibits high metabolic rates and oxygen consumption leading subsequently to a high production rate of ROS. Furthermore, inflammation, aging, genetic factors, and environmental pollution, are all elements promoting ROS generation and impairing antioxidant defense mechanisms and thereby representing risk factors leading to oxidative stress. An abnormal redox status was shown to be involved in the pathophysiology of various ocular diseases in the anterior and posterior segment of the eye. In this review, we aim to summarize the mechanisms of oxidative stress in ocular diseases to provide an updated understanding on the pathogenesis of common diseases affecting the ocular surface, the lens, the retina, and the optic nerve. Moreover, we discuss potential therapeutic approaches aimed at reducing oxidative stress in this context.
Collapse
Affiliation(s)
- Elsa Wilma Böhm
- Department of Ophthalmology, University Medical Center, Johannes Gutenberg University Mainz, Langenbeckstrasse 1, 55131, Mainz, Germany.
| | - Francesco Buonfiglio
- Department of Ophthalmology, University Medical Center, Johannes Gutenberg University Mainz, Langenbeckstrasse 1, 55131, Mainz, Germany
| | - Anna Maria Voigt
- Department of Ophthalmology, University Medical Center, Johannes Gutenberg University Mainz, Langenbeckstrasse 1, 55131, Mainz, Germany
| | - Philipp Bachmann
- Department of Ophthalmology, University Medical Center, Johannes Gutenberg University Mainz, Langenbeckstrasse 1, 55131, Mainz, Germany
| | - Tarek Safi
- Department of Ophthalmology, University Medical Center, Johannes Gutenberg University Mainz, Langenbeckstrasse 1, 55131, Mainz, Germany
| | - Norbert Pfeiffer
- Department of Ophthalmology, University Medical Center, Johannes Gutenberg University Mainz, Langenbeckstrasse 1, 55131, Mainz, Germany
| | - Adrian Gericke
- Department of Ophthalmology, University Medical Center, Johannes Gutenberg University Mainz, Langenbeckstrasse 1, 55131, Mainz, Germany.
| |
Collapse
|
91
|
Neiteler A, Palakkan AA, Gallagher KM, Ross JA. Oxidative stress and docosahexaenoic acid injury lead to increased necroptosis and ferroptosis in retinal pigment epithelium. Sci Rep 2023; 13:21143. [PMID: 38036571 PMCID: PMC10689458 DOI: 10.1038/s41598-023-47721-5] [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: 05/01/2023] [Accepted: 11/17/2023] [Indexed: 12/02/2023] Open
Abstract
Age-related macular degeneration (AMD) is a complex disease caused by different genetic and environmental risk factors leading to loss of cells in the central part of the retina. Oxidative stress appears to be an important environmental risk factor that contributes to both the initiation and progression of AMD. Retinal pigment epithelium (RPE) plays an important role in regulating oxidative stress in the retina and is one of the main retinal cell types affected in AMD. A main function of RPE is to phagocytose photoreceptor outer segments (POS) which are rich in the polyunsaturated fatty acid (PUFA) docosahexaenoic acid (DHA), making this cell type potentially more susceptible to oxidative stress-induced lipid peroxidation which can lead to cell death. RPE is known to undergo necrotic cell death in response to oxidative stress. The aim of this study was to determine if DHA in POS can increase oxidative damage to RPE. It was found that RPE undergo increased lipid peroxidation and decreased cell viability when stressed with hydrogen peroxide in combination with DHA or POS. H2O2-induced oxidative stress was found to cause both ferroptosis and necroptosis. However, the ferroptosis regulator acyl-CoA synthetase long-chain family member 4 (ACSL4) was found to be downregulated in RPE exposed to H2O2 and this effect was exacerbated when the RPE cells were simultaneously treated with DHA. Together, these results show a response of RPE when stressed which will likely be overwhelmed under disease conditions such as AMD resulting in cell death.
Collapse
Affiliation(s)
- Almar Neiteler
- Tissue Injury and Repair Group, University of Edinburgh, Chancellor's Building, 49 Little France Crescent, Edinburgh, EH16 4SB, UK.
| | - Anwar A Palakkan
- Immunology and Stem Cell Biology, Aravind Medical Research Foundation, Anna Nagar, Madurai, 625020, India
| | - Kevin M Gallagher
- Tissue Injury and Repair Group, University of Edinburgh, Chancellor's Building, 49 Little France Crescent, Edinburgh, EH16 4SB, UK
| | - James A Ross
- Tissue Injury and Repair Group, University of Edinburgh, Chancellor's Building, 49 Little France Crescent, Edinburgh, EH16 4SB, UK
| |
Collapse
|
92
|
Ijurko C, Romo-González M, García-Calvo C, Sardina JL, Sánchez-Bernal C, Sánchez-Yagüe J, Elena-Herrmann B, Villaret J, Garrel C, Mondet J, Mossuz P, Hernández-Hernández Á. NOX2 control over energy metabolism plays a role in acute myeloid leukaemia prognosis and survival. Free Radic Biol Med 2023; 209:18-28. [PMID: 37806599 DOI: 10.1016/j.freeradbiomed.2023.10.013] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Revised: 10/03/2023] [Accepted: 10/04/2023] [Indexed: 10/10/2023]
Abstract
Acute myeloid leukaemia (AML) is a highly heterogeneous disease, however the therapeutic approaches have hardly changed in the last decades. Metabolism rewiring and the enhanced production of reactive oxygen species (ROS) are hallmarks of cancer. A deeper understanding of these features could be instrumental for the development of specific AML-subtypes treatments. NADPH oxidases (NOX), the only cellular system specialised in ROS production, are also involved in leukemic metabolism control. NOX2 shows a variable expression in AML patients, so patients can be classified based on such difference. Here we have analysed whether NOX2 levels are important for AML metabolism control. The lack of NOX2 in AML cells slowdowns basal glycolysis and oxidative phosphorylation (OXPHOS), along with the accumulation of metabolites that feed such routes, and a sharp decrease of glutathione. In addition, we found changes in the expression of 725 genes. Among them, we have discovered a panel of 30 differentially expressed metabolic genes, whose relevance was validated in patients. This panel can segregate AML patients according to CYBB expression, and it can predict patient prognosis and survival. In summary, our data strongly support the relevance of NOX2 for AML metabolism, and highlights the potential of our discoveries in AML prognosis.
Collapse
Affiliation(s)
- Carla Ijurko
- Departamento de Bioquímica y Biología Molecular, Universidad de Salamanca, Salamanca, 37007, Spain; IBSAL (Instituto de Investigación Biomédica de Salamanca), Salamanca, 37007, Spain
| | - Marta Romo-González
- Departamento de Bioquímica y Biología Molecular, Universidad de Salamanca, Salamanca, 37007, Spain; IBSAL (Instituto de Investigación Biomédica de Salamanca), Salamanca, 37007, Spain
| | - Clara García-Calvo
- Departamento de Bioquímica y Biología Molecular, Universidad de Salamanca, Salamanca, 37007, Spain; IBSAL (Instituto de Investigación Biomédica de Salamanca), Salamanca, 37007, Spain
| | - José Luis Sardina
- Epigenetic Control of Haematopoiesis Group, Josep Carreras Leukaemia Research Institute, Barcelona, Spain
| | - Carmen Sánchez-Bernal
- Departamento de Bioquímica y Biología Molecular, Universidad de Salamanca, Salamanca, 37007, Spain; IBSAL (Instituto de Investigación Biomédica de Salamanca), Salamanca, 37007, Spain
| | - Jesús Sánchez-Yagüe
- Departamento de Bioquímica y Biología Molecular, Universidad de Salamanca, Salamanca, 37007, Spain; IBSAL (Instituto de Investigación Biomédica de Salamanca), Salamanca, 37007, Spain
| | - Bénédicte Elena-Herrmann
- University Grenoble Alpes, Inserm U 1209, CNRS UMR 5309, GEMELI Platform, Institute for Advanced Biosciences, 38000, Grenoble, France
| | - Joran Villaret
- University Grenoble Alpes, Inserm U 1209, CNRS UMR 5309, GEMELI Platform, Institute for Advanced Biosciences, 38000, Grenoble, France
| | - Catherine Garrel
- Department of Biochemistry, Institute of Biology and Pathology, Hospital of Grenoble Alpes (CHUGA), CS 20217, 38043, Grenoble, CEDEX 9, France
| | - Julie Mondet
- Team "Epigenetic Regulations", Institute for Advanced Biosciences, University Grenoble Alpes (UGA), INSERM U1209/CNRS 5309, 38700, Grenoble, France; Department of Molecular Pathology, Institute of Biology and Pathology, Hospital of Grenoble Alpes (CHUGA), CS 20217, 38043, Grenoble, CEDEX 9, France
| | - Pascal Mossuz
- Team "Epigenetic Regulations", Institute for Advanced Biosciences, University Grenoble Alpes (UGA), INSERM U1209/CNRS 5309, 38700, Grenoble, France; Department of Biological Hematology, Institute of Biology and Pathology, Hospital of Grenoble Alpes (CHUGA), CS 20217, 38043, Grenoble, CEDEX 9, France
| | - Ángel Hernández-Hernández
- Departamento de Bioquímica y Biología Molecular, Universidad de Salamanca, Salamanca, 37007, Spain; IBSAL (Instituto de Investigación Biomédica de Salamanca), Salamanca, 37007, Spain.
| |
Collapse
|
93
|
Chhunchha B, Kumar R, Kubo E, Thakur P, Singh DP. Prdx6 Regulates Nlrp3 Inflammasome Activation-Driven Inflammatory Response in Lens Epithelial Cells. Int J Mol Sci 2023; 24:16276. [PMID: 38003466 PMCID: PMC10671722 DOI: 10.3390/ijms242216276] [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: 10/15/2023] [Revised: 11/06/2023] [Accepted: 11/08/2023] [Indexed: 11/26/2023] Open
Abstract
The continuum of antioxidant response dysregulation in aging/oxidative stress-driven Nlrp3 inflammasome activation-mediated inflammatory response is associated with age-related diseases. Peroxiredoxin (Prdx) 6 is a key antioxidant that provides cytoprotection by regulating redox homeostasis. Herein, using lens epithelial cells (LECs) derived from the targeted inactivation of Prdx6 gene and aging lenses, we present molecular evidence that Prdx6-deficiency causes oxidative-driven Nlrp3 inflammasome activation, resulting in pyroptosis in aging/redox active cells wherein Prdx6 availability offsets the inflammatory process. We observed that Prdx6-/- and aging LECs harboring accumulated reactive oxygen species (ROS) showed augmented activation of Nlrp3 and bioactive inflammatory components, like Caspase-1, IL-1β, ASC and Gasdermin-D. Similar to lipopolysaccharide treatment, oxidative exposure led to further ROS amplification with increased activation of the Nlrp3 inflammasome pathway. Mechanistically, we found that oxidative stress enhanced Kruppel-like factor 9 (Klf9) expression in aging/Prdx6-/- mLECs, leading to a Klf9-dependent increase in Nlrp3 transcription, while the elimination of ROS by the delivery of Prdx6 or by silencing Klf9 prevented the inflammatory response. Altogether, our data identify the biological significance of Prdx6 as an intrinsic checkpoint for regulating the cellular health of aging or redox active LECs and provide opportunities to develop antioxidant-based therapeutic(s) to prevent oxidative/aging-related diseases linked to aberrant Nlrp3 inflammasome activation.
Collapse
Affiliation(s)
- Bhavana Chhunchha
- Department of Ophthalmology and Visual Sciences, University of Nebraska Medical Center, Omaha, NE 68198, USA; (R.K.); (P.T.)
| | - Rakesh Kumar
- Department of Ophthalmology and Visual Sciences, University of Nebraska Medical Center, Omaha, NE 68198, USA; (R.K.); (P.T.)
| | - Eri Kubo
- Department of Ophthalmology, Kanazawa Medical University, Kahoku 9200293, Ishikawa, Japan;
| | - Priyanka Thakur
- Department of Ophthalmology and Visual Sciences, University of Nebraska Medical Center, Omaha, NE 68198, USA; (R.K.); (P.T.)
| | - Dhirendra P. Singh
- Department of Ophthalmology and Visual Sciences, University of Nebraska Medical Center, Omaha, NE 68198, USA; (R.K.); (P.T.)
| |
Collapse
|
94
|
Tranah GJ, Barnes HN, Cawthon PM, Coen PM, Esser KA, Hepple RT, Huo Z, Kramer PA, Toledo FGS, Evans DS, Cummings SR. Expression of mitochondrial oxidative stress response genes in muscle is associated with mitochondrial respiration, physical performance, and muscle mass in the Study of Muscle, Mobility and Aging (SOMMA). MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2023:2023.11.05.23298108. [PMID: 37986804 PMCID: PMC10659517 DOI: 10.1101/2023.11.05.23298108] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/22/2023]
Abstract
Gene expression in skeletal muscle of older individuals may reflect compensatory adaptations in response to oxidative damage that preserve tissue integrity and maintain function. Identifying associations between oxidative stress response gene expression patterns and mitochondrial function, physical performance, and muscle mass in older individuals would further our knowledge of mechanisms related to managing molecular damage that may be targeted to preserve physical resilience. To characterize expression patterns of genes responsible for the oxidative stress response, RNA was extracted and sequenced from skeletal muscle biopsies collected from 575 participants (≥70 years old) from the Study of Muscle, Mobility and Aging. Expression levels of twenty-one protein coding RNAs related to the oxidative stress response were analyzed in relation to six phenotypic measures, including: maximal mitochondrial respiration from muscle biopsies (Max OXPHOS), physical performance (VO2 peak, 400m walking speed, and leg strength), and muscle size (thigh muscle volume and whole-body D3Cr muscle mass). The mRNA level of the oxidative stress response genes most consistently associated across outcomes are preferentially expressed within the mitochondria. Higher expression of mRNAs that encode generally mitochondria located proteins SOD2, TRX2, PRX3, PRX5, and GRX2 were associated with higher levels of mitochondrial respiration and VO2 peak. In addition, greater SOD2, PRX3, and GRX2 expression was associated with higher physical performance and muscle size. Identifying specific mechanisms associated with high functioning across multiple performance and physical domains may lead to targeted antioxidant interventions with greater impacts on mobility and independence.
Collapse
Affiliation(s)
- Gregory J Tranah
- California Pacific Medical Center Research Institute, San Francisco, California, USA
- Department of Epidemiology and Biostatistics, University of California San Francisco, San Francisco, California, USA
| | - Haley N Barnes
- California Pacific Medical Center Research Institute, San Francisco, California, USA
| | - Peggy M Cawthon
- California Pacific Medical Center Research Institute, San Francisco, California, USA
- Department of Epidemiology and Biostatistics, University of California San Francisco, San Francisco, California, USA
| | - Paul M Coen
- Translational Research Institute, Advent Health, Orlando, Florida, USA
| | - Karyn A Esser
- Department of Physiology and Ageing, College of Medicine, University of Florida, Gainesville, FL, USA
| | - Russell T Hepple
- Department of Physical Therapy, University of Florida, Gainesville, Florida, USA
| | - Zhiguang Huo
- Department of Biostatistics, College of Public Health & Health Professions, College of Medicine University of Florida, Gainesville, Florida, USA
| | - Philip A Kramer
- Department of Internal Medicine-Gerontology and Geriatric Medicine, Wake Forest University School of Medicine, Winston-Salem, North Carolina, USA
| | - Frederico G S Toledo
- Division of Endocrinology and Metabolism, Department of Medicine, University of Pittsburgh, USA
| | - Daniel S Evans
- California Pacific Medical Center Research Institute, San Francisco, California, USA
- Department of Epidemiology and Biostatistics, University of California San Francisco, San Francisco, California, USA
| | - Steven R Cummings
- California Pacific Medical Center Research Institute, San Francisco, California, USA
- Department of Epidemiology and Biostatistics, University of California San Francisco, San Francisco, California, USA
| |
Collapse
|
95
|
Fan JB, Yuan K, Zhu XH, Cui SY, Yi H, Zhang W. Neuroligin-3 activates Akt-dependent Nrf2 cascade to protect osteoblasts from oxidative stress. Free Radic Biol Med 2023; 208:807-819. [PMID: 37774803 DOI: 10.1016/j.freeradbiomed.2023.09.032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Revised: 09/24/2023] [Accepted: 09/26/2023] [Indexed: 10/01/2023]
Abstract
Excessive oxidative stress will cause significant injury to osteoblasts, serving as one major pathological mechanism of osteoporosis. Neuroligin-3 (NLGN3) is a postsynaptic cell adhesion protein and is expressed in the bone. We here explored its potential activity against hydrogen peroxide (H2O2)-induced oxidative injury in cultured osteoblasts. In primary murine and human osteoblasts, NLGN3 stimulation dose-dependently induced Akt, Erk1/2 and S6K activation. NLGN3 pretreatment ameliorated H2O2-induced cytotoxicity and death in osteoblasts. Moreover, H2O2-induced reactive oxygen species (ROS) production and oxidative injury were alleviated with NLGN3 pretreatment in cultured osteoblasts. Further studies showed that NLGN3 activated Nrf2 signaling cascade and induced Nrf2 protein Serine-40 phosphorylation, Keap1-Nrf2 dissociation, Nrf2 protein stabilization and nuclear translocation in osteoblasts. NLGN3 also increased antioxidant response element (ARE) activity and induced expression of Nrf2-ARE-dependent genes (HO1, GCLC and NQO1) in osteoblasts. Moreover NLGN3 mitigated osteoblast oxidative injury by dexamethasone or sodium fluoride (NaF). Nrf2 cascade activation is essential for NLGN3-induced cytoprotective activity in osteoblasts. Nrf2 shRNA or knockout (KO) abolished NLGN3-induced osteoblast cytoprotection against H2O2. Contrarily forced Nrf2 cascade activation by Keap1 KO mimicked NLGN3-induced anti-oxidative activity in murine osteoblasts. Importantly, NLGN3-induced Serine-40 phosphorylation and Nrf2 cascade activation were blocked by an Akt inhibitor MK-2206 or by Akt1 shRNA. Importantly, Akt inhibition, Akt1 silencing or Nrf2 S40T mutation largely inhibited NLGN3-induced osteoblast cytoprotection against H2O2. At last, we showed that NLGN3 mRNA and protein expression was significantly downregulated in necrotic bone tissues of dexamethasone-taken patients. Taken together, NLGN3 activated Akt-dependent Nrf2 cascade to protect osteoblasts from oxidative stress.
Collapse
Affiliation(s)
- Jian-Bo Fan
- Department of Orthopaedics, Affiliated Hospital 2 of Nantong University, Nantong, China.
| | - Kun Yuan
- Department of Orthopaedics, Affiliated Hospital 2 of Nantong University, Nantong, China
| | - Xin-Hui Zhu
- Department of Orthopaedics, Affiliated Hospital 2 of Nantong University, Nantong, China
| | - Sheng-Yu Cui
- Department of Orthopaedics, Affiliated Hospital 2 of Nantong University, Nantong, China
| | - Hong Yi
- Department of Orthopaedics, Affiliated Hospital 2 of Nantong University, Nantong, China
| | - Wei Zhang
- Department of Orthopaedics, Affiliated Hospital 2 of Nantong University, Nantong, China
| |
Collapse
|
96
|
Reich H, Savage-Dunn C. Signaling circuits and the apical extracellular matrix in aging: connections identified in the nematode Caenorhabditis elegans. Am J Physiol Cell Physiol 2023; 325:C1201-C1211. [PMID: 37721005 PMCID: PMC10861026 DOI: 10.1152/ajpcell.00195.2023] [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: 05/09/2023] [Revised: 08/24/2023] [Accepted: 09/11/2023] [Indexed: 09/19/2023]
Abstract
Numerous conserved signaling pathways play critical roles in aging, including insulin/IGF-1, TGF-β, and Wnt pathways. Some of these pathways also play prominent roles in the formation and maintenance of the extracellular matrix. The nematode Caenorhabditis elegans has been an enduringly productive system for the identification of conserved mechanisms of biological aging. Recent studies in C. elegans highlight the regulatory circuits between conserved signaling pathways and the extracellular matrix, revealing a bidirectional relationship between these factors and providing a platform to address how regulation of and by the extracellular matrix can impact lifespan and organismal health during aging. These discoveries provide new opportunities for clinical advances and novel therapeutic strategies.
Collapse
Affiliation(s)
- Hannah Reich
- Department of Biology, Queens College, City University of New York, Flushing, New York, United States
| | - Cathy Savage-Dunn
- Department of Biology, Queens College, City University of New York, Flushing, New York, United States
- PhD Program in Biology, The Graduate Center, City University of New York, New York, New York, United States
| |
Collapse
|
97
|
He SY, Liu RP, Wang CR, Wang XQ, Wang J, Xu YN, Kim NH, Han DW, Li YH. Improving the developmental competences of porcine parthenogenetic embryos by Notoginsenoside R1-induced enhancement of mitochondrial activity and alleviation of proapoptotic events. Reprod Domest Anim 2023; 58:1583-1594. [PMID: 37696770 DOI: 10.1111/rda.14474] [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: 05/13/2023] [Revised: 08/08/2023] [Accepted: 09/01/2023] [Indexed: 09/13/2023]
Abstract
Notoginsenoside R1 (NGR1), derived from the Panax notoginseng root and rhizome, exhibits diverse pharmacological influences on the brain, neurons, and osteoblasts, such as antioxidant effects, mitochondrial function protection, energy metabolism regulation, and inhibition of oxygen radicals, apoptosis, and cellular autophagy. However, its effect on early porcine embryonic development remains unclear. Therefore, we investigated NGR1's effects on blastocyst quality, reactive oxygen species (ROS) levels, glutathione (GSH) levels, mitochondrial function, and embryonic development-related gene expression in porcine embryos by introducing NGR1 during the in vitro culture (IVC) of early porcine embryos. Our results indicate that an addition of 1 μM NGR1 significantly increased glutathione (GSH) levels, blastocyst formation rate, and total cell number and proliferation capacity; decreased ROS levels and apoptosis rates in orphan-activated porcine embryos; and improved intracellular mitochondrial distribution, enhanced membrane potential, and reduced autophagy. In addition, pluripotency-related factor levels were elevated (NANOG and octamer-binding transcription factor 4 [OCT4]), antioxidant-related genes were upregulated (nuclear factor-erythroid 2-related factor 2 [NRF2]), and apoptosis- (caspase 3 [CAS3]) and autophagy-related genes (light chain 3 [LC3B]) were downregulated. These results indicate that NGR1 can enhance early porcine embryonic development by protecting mitochondrial function.
Collapse
Affiliation(s)
- Sheng-Yan He
- Guangdong Provincial Key Laboratory of Large Animal Models for Biomedicine, School of Biotechnology and Health Sciences, Wuyi University, Jiangmen, China
| | - Rong-Ping Liu
- Guangdong Provincial Key Laboratory of Large Animal Models for Biomedicine, School of Biotechnology and Health Sciences, Wuyi University, Jiangmen, China
| | - Chao-Rui Wang
- Guangdong Provincial Key Laboratory of Large Animal Models for Biomedicine, School of Biotechnology and Health Sciences, Wuyi University, Jiangmen, China
| | - Xin-Qin Wang
- Guangdong Provincial Key Laboratory of Large Animal Models for Biomedicine, School of Biotechnology and Health Sciences, Wuyi University, Jiangmen, China
| | - Jing Wang
- Guangdong Provincial Key Laboratory of Large Animal Models for Biomedicine, School of Biotechnology and Health Sciences, Wuyi University, Jiangmen, China
| | - Yong-Nan Xu
- Guangdong Provincial Key Laboratory of Large Animal Models for Biomedicine, School of Biotechnology and Health Sciences, Wuyi University, Jiangmen, China
| | - Nam-Hyung Kim
- Guangdong Provincial Key Laboratory of Large Animal Models for Biomedicine, School of Biotechnology and Health Sciences, Wuyi University, Jiangmen, China
| | - Dong-Wook Han
- Guangdong Provincial Key Laboratory of Large Animal Models for Biomedicine, School of Biotechnology and Health Sciences, Wuyi University, Jiangmen, China
| | - Ying-Hua Li
- Guangdong Provincial Key Laboratory of Large Animal Models for Biomedicine, School of Biotechnology and Health Sciences, Wuyi University, Jiangmen, China
| |
Collapse
|
98
|
Shastak Y, Pelletier W. Captivating Colors, Crucial Roles: Astaxanthin's Antioxidant Impact on Fish Oxidative Stress and Reproductive Performance. Animals (Basel) 2023; 13:3357. [PMID: 37958112 PMCID: PMC10648254 DOI: 10.3390/ani13213357] [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: 09/06/2023] [Revised: 10/23/2023] [Accepted: 10/26/2023] [Indexed: 11/15/2023] Open
Abstract
Fish, constantly exposed to environmental stressors due to their aquatic habitat and high metabolic rates, are susceptible to oxidative stress. This review examines the interplay between oxidative stress and fish reproduction, emphasizing the potent antioxidant properties of astaxanthin. Our primary objective is to highlight astaxanthin's role in mitigating oxidative stress during critical reproductive stages, leading to improved gamete quality, ovary development, and hormone levels. We also explore its practical applications in aquaculture, including enhanced pigmentation and overall fish health. We conducted a comprehensive literature review, analyzing studies on astaxanthin's antioxidant properties and its impact on fish reproduction. Astaxanthin, a carotenoid pigment, effectively combats reactive oxygen species, inhibiting lipid peroxidation and maintaining membrane integrity. It significantly enhances reproductive success in fish and improves overall fish health in aquaculture settings. This review reveals astaxanthin's multifaceted benefits in fish health and reproduction, offering economic advantages in aquaculture. Future research should delve into species-specific responses, optimal dosages, and the long-term effects of astaxanthin supplementation to inform sustainable aquaculture strategies.
Collapse
Affiliation(s)
- Yauheni Shastak
- Nutrition & Health Division, BASF SE, 67063 Ludwigshafen am Rhein, Germany
| | | |
Collapse
|
99
|
Chen Z, Wang P, Cheng H, Wang N, Wu M, Wang Z, Wang Z, Dong W, Guan D, Wang L, Zhao R. Adolescent traumatic brain injury leads to incremental neural impairment in middle-aged mice: role of persistent oxidative stress and neuroinflammation. Front Neurosci 2023; 17:1292014. [PMID: 37965213 PMCID: PMC10642192 DOI: 10.3389/fnins.2023.1292014] [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: 09/10/2023] [Accepted: 10/13/2023] [Indexed: 11/16/2023] Open
Abstract
BACKGROUND Traumatic brain injury (TBI) increases the risk of mental disorders and neurodegenerative diseases in the chronic phase. However, there is limited neuropathological or molecular data on the long-term neural dysfunction and its potential mechanism following adolescent TBI. METHODS A total of 160 male mice aged 8 weeks were used to mimic moderate TBI by controlled cortical impact. At 1, 3, 6 and 12 months post-injury (mpi), different neurological functions were evaluated by elevated plus maze, forced swimming test, sucrose preference test and Morris water maze. The levels of oxidative stress, antioxidant response, reactive astrocytes and microglia, and expression of inflammatory cytokines were subsequently assessed in the ipsilateral hippocampus, followed by neuronal apoptosis detection. Additionally, the morphological complexity of hippocampal astrocytes was evaluated by Sholl analysis. RESULTS The adolescent mice exhibited persistent and incremental deficits in memory and anxiety-like behavior after TBI, which were sharply exacerbated at 12 mpi. Depression-like behaviors were observed in TBI mice at 6 mpi and 12 mpi. Compared with the age-matched control mice, apoptotic neurons were observed in the ipsilateral hippocampus during the chronic phase of TBI, which were accompanied by enhanced oxidative stress, and expression of inflammatory cytokines (IL-1β and TNF-α). Moreover, the reactive astrogliosis and microgliosis in the ipsilateral hippocampus were observed in the late phase of TBI, especially at 12 mpi. CONCLUSION Adolescent TBI leads to incremental cognitive dysfunction, and depression- and anxiety-like behaviors in middle-aged mice. The chronic persistent neuroinflammation and oxidative stress account for the neuronal loss and neural dysfunction in the ipsilateral hippocampus. Our results provide evidence for the pathogenesis of chronic neural damage following TBI and shed new light on the treatment of TBI-induced late-phase neurological dysfunction.
Collapse
Affiliation(s)
- Ziyuan Chen
- Department of Forensic Pathology, School of Forensic Medicine, China Medical University, Shenyang, Liaoning, China
| | - Pengfei Wang
- Department of Forensic Pathology, School of Forensic Medicine, China Medical University, Shenyang, Liaoning, China
| | - Hao Cheng
- Department of Forensic Pathology, School of Forensic Medicine, China Medical University, Shenyang, Liaoning, China
| | - Ning Wang
- Department of Forensic Pathology, School of Forensic Medicine, China Medical University, Shenyang, Liaoning, China
| | - Mingzhe Wu
- Department of Forensic Pathology, School of Forensic Medicine, China Medical University, Shenyang, Liaoning, China
| | - Ziwei Wang
- Department of Forensic Pathology, School of Forensic Medicine, China Medical University, Shenyang, Liaoning, China
| | - Zhi Wang
- Department of Forensic Pathology, School of Forensic Medicine, China Medical University, Shenyang, Liaoning, China
| | - Wenwen Dong
- Department of Forensic Pathology, School of Forensic Medicine, China Medical University, Shenyang, Liaoning, China
| | - Dawei Guan
- Department of Forensic Pathology, School of Forensic Medicine, China Medical University, Shenyang, Liaoning, China
| | - Linlin Wang
- Department of Forensic Pathology, School of Forensic Medicine, China Medical University, Shenyang, Liaoning, China
| | - Rui Zhao
- Department of Forensic Pathology, School of Forensic Medicine, China Medical University, Shenyang, Liaoning, China
- Key Laboratory of Environmental Stress and Chronic Disease Control and Prevention, Ministry of Education, China Medical University, Shenyang, Liaoning, China
- Liaoning Province Key Laboratory of Forensic Bio-Evidence Sciences, Shenyang, China
| |
Collapse
|
100
|
Han Y, Liu Y, Zhang Y, Wang W, Lv T, Huang J, Peng X. The Role and Application of the AMPK-Sirtuins Network in Cellular Senescence. FRONT BIOSCI-LANDMRK 2023; 28:250. [PMID: 37919064 DOI: 10.31083/j.fbl2810250] [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: 04/03/2023] [Revised: 05/22/2023] [Accepted: 05/26/2023] [Indexed: 11/04/2023]
Abstract
Aging and related diseases significantly affect the health and happiness index around the world. Cellular senescence is the basis of physiological aging and is closely related to various senile diseases. AMP-activated protein kinase (AMPK) is associated with both the regulation of cellular energy metabolism and the regulation of cellular senescence. Another set of proteins, sirtuins, has also been demonstrated to play an important role in cell senescence. However, it is not clear how AMPK and sirtuins coordinate to regulate cellular senescence. Herein, we summarized the role of AMPK and sirtuins in regulating metabolism, repairing DNA damage, and even prolonging human life. We have provided a detailed explanation of the clinical trials relating to the AMPK and sirtuins involved in aging. Systematically analyzing individual senescence genes and developing functional reference notes will aid in understanding the potential mechanisms underlying aging and identify therapeutic targets for both anti-aging interventions and age-related illnesses.
Collapse
Affiliation(s)
- Yukun Han
- PET Center of Nuclear Medicine, Department of the First Affiliated Hospital of Yangtze University, and School of Medicine of Yangtze University, 434000 Jingzhou, Hubei, China
- Department of Pathophysiology, School of Basic Medicine, Health Science Center, Yangtze University, 434023 Jingzhou, Hubei, China
| | - Yifan Liu
- Department of Pathophysiology, School of Basic Medicine, Health Science Center, Yangtze University, 434023 Jingzhou, Hubei, China
- Department of Oncology, Jingzhou Hospital Affifiliated to Yangtze University, 434023 Jingzhou, Hubei, China
| | - Yanhua Zhang
- Department of Pathophysiology, School of Basic Medicine, Health Science Center, Yangtze University, 434023 Jingzhou, Hubei, China
- Laboratory of Oncology, Center for Molecular Medicine, School of Basic Medicine, Health Science Center, Yangtze University, 434023 Jingzhou, Hubei, China
| | - Wei Wang
- Department of Rehabilitation Radiology, Beijing Rehabilitation Hospital, Capital Medical University, 100144 Beijing, China
| | - Tongtong Lv
- Department of Pathophysiology, School of Basic Medicine, Health Science Center, Yangtze University, 434023 Jingzhou, Hubei, China
- Laboratory of Oncology, Center for Molecular Medicine, School of Basic Medicine, Health Science Center, Yangtze University, 434023 Jingzhou, Hubei, China
| | - Jinbai Huang
- PET Center of Nuclear Medicine, Department of the First Affiliated Hospital of Yangtze University, and School of Medicine of Yangtze University, 434000 Jingzhou, Hubei, China
| | - Xiaochun Peng
- Department of Pathophysiology, School of Basic Medicine, Health Science Center, Yangtze University, 434023 Jingzhou, Hubei, China
- Laboratory of Oncology, Center for Molecular Medicine, School of Basic Medicine, Health Science Center, Yangtze University, 434023 Jingzhou, Hubei, China
| |
Collapse
|