1
|
Liang J, Yang F, Li Z, Li Q. Epigenetic regulation of the inflammatory response in stroke. Neural Regen Res 2025; 20:3045-3062. [PMID: 39589183 DOI: 10.4103/nrr.nrr-d-24-00672] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Accepted: 09/20/2024] [Indexed: 11/27/2024] Open
Abstract
Stroke is classified as ischemic or hemorrhagic, and there are few effective treatments for either type. Immunologic mechanisms play a critical role in secondary brain injury following a stroke, which manifests as cytokine release, blood-brain barrier disruption, neuronal cell death, and ultimately behavioral impairment. Suppressing the inflammatory response has been shown to mitigate this cascade of events in experimental stroke models. However, in clinical trials of anti-inflammatory agents, long-term immunosuppression has not demonstrated significant clinical benefits for patients. This may be attributable to the dichotomous roles of inflammation in both tissue injury and repair, as well as the complex pathophysiologic inflammatory processes in stroke. Inhibiting acute harmful inflammatory responses or inducing a phenotypic shift from a pro-inflammatory to an anti-inflammatory state at specific time points after a stroke are alternative and promising therapeutic strategies. Identifying agents that can modulate inflammation requires a detailed understanding of the inflammatory processes of stroke. Furthermore, epigenetic reprogramming plays a crucial role in modulating post-stroke inflammation and can potentially be exploited for stroke management. In this review, we summarize current findings on the epigenetic regulation of the inflammatory response in stroke, focusing on key signaling pathways including nuclear factor-kappa B, Janus kinase/signal transducer and activator of transcription, and mitogen-activated protein kinase as well as inflammasome activation. We also discuss promising molecular targets for stroke treatment. The evidence to date indicates that therapeutic targeting of the epigenetic regulation of inflammation can shift the balance from inflammation-induced tissue injury to repair following stroke, leading to improved post-stroke outcomes.
Collapse
Affiliation(s)
- Jingyi Liang
- School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Fei Yang
- Department of Neurobiology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
- Laboratory for Clinical Medicine, Beijing Key Laboratory of Neural Regeneration and Repair, Capital Medical University, Beijing, China
| | - Zixiao Li
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- China National Clinical Research Center for Neurological Diseases, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- National Center for Healthcare Quality Management in Neurological Diseases, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- Chinese Institute for Brain Research, Beijing, China
- Research Unit of Artificial Intelligence in Cerebrovascular Disease, Chinese Academy of Medical Sciences, Beijing, China
- Beijing Engineering Research Center of Digital Healthcare for Neurological Diseases, Beijing, China
| | - Qian Li
- Laboratory for Clinical Medicine, Beijing Key Laboratory of Neural Regeneration and Repair, Capital Medical University, Beijing, China
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
- Beijing Key Laboratory of Cancer Invasion and Metastasis Research, Capital Medical University, Beijing, China
| |
Collapse
|
2
|
Park JW, Park JH, Lee H, Wang C, Cao S, Kim HS. Penilumamide, a novel SIRT1 activator, protects UVB-induced photodamages in HaCaT cells. JOURNAL OF TOXICOLOGY AND ENVIRONMENTAL HEALTH. PART A 2025; 88:106-121. [PMID: 39714391 DOI: 10.1080/15287394.2024.2387041] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2024]
Abstract
Ultraviolet-B (UVB) radiation is a major physical factor that induces structural changes in human skin. The aim of this study was to determine whether the novel silent information regulator 1 (sirtuin 1 SIRT1) protein activator, penilumamide, exerted any protective effects against UVB-induced skin damage using human HaCaT keratinocytes as a model. Enzymatic assays were performed to determine the SIRT1-activating ability of penilumamide, which was compared with that of resveratrol, a potent natural product SIRT1 activator with antioxidant and anti-inflammatory properties. Penilumamide markedly activated SIRT1 enzyme activity compared to resveratrol. To further investigate the protective effect of penilumamide against UVB-induced cytotoxicity, HaCaT cells were pretreated with penilumamide (10 μM) for 24 hr followed by irradiation with UVB (40 mJ/cm2). UVB (40 mJ/cm2) irradiation significantly reduced cell viability in a time-dependent manner, whereas pretreatment with penilumamide blocked this effect. Further, penilumamide decreased the levels of intracellular reactive oxygen species (ROS) generated by UVB irradiation in HaCaT cells. Pretreatment with penilumamide also prevented UVB irradiation-induced changes in mitochondrial membrane potential (ΔΨm). In addition, pretreatment with penilumamide significantly reduced the expression levels of pro-inflammatory cytokines, interleukin (IL)-6, IL-8, and IL-10 and phosphorylation of nuclear factor-kB (NF-kB). These results indicate that penilumamide protects HaCaT cells from UVB-induced inflammation. Taken together data demonstrate that penilumamide exerted protective effects against UVB-induced ROS generation in HaCaT cells. Therefore, penilumamide may be considered to be used as a new SIRT1 activator to protect human keratinocyte against UVB-induced damage.
Collapse
Affiliation(s)
- Ji Won Park
- School of Pharmacy, Sungkyunkwan University, Suwon, Gyeonggi-do, Republic of Korea
| | - Jae Hyeon Park
- School of Pharmacy, Sungkyunkwan University, Suwon, Gyeonggi-do, Republic of Korea
| | - Haeun Lee
- School of Pharmacy, Sungkyunkwan University, Suwon, Gyeonggi-do, Republic of Korea
| | - Cong Wang
- Department of Pharmaceutical Sciences, Daniel K. Inouye College of Pharmacy, University of Hawai'i, Hilo, HI, USA
| | - Shugeng Cao
- Department of Pharmaceutical Sciences, Daniel K. Inouye College of Pharmacy, University of Hawai'i, Hilo, HI, USA
| | - Hyung Sik Kim
- School of Pharmacy, Sungkyunkwan University, Suwon, Gyeonggi-do, Republic of Korea
| |
Collapse
|
3
|
Ghaith WZ, Wadie W, El-Yamany MF. Crosstalk between SIRT1/Nrf2 signaling and NLRP3 inflammasome/pyroptosis as a mechanistic approach for the neuroprotective effect of linagliptin in Parkinson's disease. Int Immunopharmacol 2025; 145:113716. [PMID: 39642562 DOI: 10.1016/j.intimp.2024.113716] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2024] [Revised: 11/24/2024] [Accepted: 11/24/2024] [Indexed: 12/09/2024]
Abstract
In recent years, special attention has been paid to highlighting the antiparkinsonian effect of linagliptin. However, the mechanism of its action has not yet been well investigated. The present study aimed to verify the neuroprotective effect of linagliptin in the rotenone model of Parkinson's disease (PD) and further explore its potential molecular mechanisms. Rats were intoxicated with rotenone (2 mg/kg/day; sc) and treated with linagliptin (10 mg/kg/day; po) for 14 consecutive days. The present finding showed that linagliptin ameliorated the histopathological changes of rotenone on substantia nigra and striata. Linagliptin decreased α-synuclein immunoreactivity along with an increase in tyrosine hydroxylase immunoreactivity and striatal dopamine content. This was reflected in the marked improvement of the behavior and motor deficits in rotenone-intoxicated rats. On the molecular level, linagliptin upregulated sirtuin 1 (SIRT1)/ nuclear factor erythroid 2-related factor 2 (Nrf2) signaling, reduced ionized calcium-binding adaptor molecule 1 (Iba1) protein expression, restored glutathione (GSH) content, and elevated heme oxygenase-1 (HO-1) level in rats with rotenone intoxication. Moreover, linagliptin inhibited NOD-like receptor protein 3 (NLRP3)/caspase-1/interleukin-1β (IL-1β) cascade with subsequent reduction in gasdermin D (GSDMD) expression. Therefore, the present study reveals the ability of linagliptin, through the activation of SIRT1/Nrf2 signaling, to suppress NLRP3 inflammasome-mediated pyroptosis and protect against rotenone-induced parkinsonism.
Collapse
Affiliation(s)
| | - Walaa Wadie
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, 11562, Egypt
| | - Mohammed F El-Yamany
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, 11562, Egypt.
| |
Collapse
|
4
|
Placke JM, Bottek J, Váraljai R, Shannan B, Scharfenberg S, Krisp C, Spangenberg P, Soun C, Siemes D, Borgards L, Hoffmann F, Zhao F, Paschen A, Schlueter H, von Eggeling F, Helfrich I, Rambow F, Ugurel S, Tasdogan A, Schadendorf D, Engel DR, Roesch A. Spatial proteomics reveals sirtuin 1 to be a determinant of T-cell infiltration in human melanoma. Br J Dermatol 2024:ljae433. [PMID: 39739311 DOI: 10.1093/bjd/ljae433] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Revised: 10/30/2024] [Indexed: 01/02/2025]
Abstract
BACKGROUND The tumour microenvironment significantly influences the clinical response of patients to therapeutic immune checkpoint inhibition (ICI), but a comprehensive understanding of the underlying immune-regulatory proteome is still lacking. OBJECTIVES To decipher targetable biologic processes that determine tumour-infiltrating lymphocytes (TiLs) as a cellular equivalent of clinical response to ICI. METHODS We mapped the spatial distribution of proteins in TiL-enriched vs. TiL-low compartments in melanoma by combining microscopy, matrix-assisted laser desorption mass spectrometry imaging and liquid chromatography-mass spectrometry, as well as computational data mining. Pharmacological modulation of sirtuin 1 (SIRT1) activity in syngeneic mouse models was used to evaluate the efficacy of pharmacological SIRT1 activation in two syngeneic melanoma mouse models, one known to be α-programmed cell death protein 1 (PD-1) sensitive and the other α-PD-1 resistant. RESULTS Spatial proteomics and gene ontology-based enrichment analysis identified > 145 proteins enriched in CD8high tumour compartments, including negative regulators of mammalian target of rapamycin signalling such as SIRT1. Multiplexed immunohistochemistry confirmed that SIRT1 protein was expressed more in CD8high than in CD8low compartments. Further analysis of bulk and single-cell RNA sequencing data from melanoma tissue samples suggested the expression of SIRT1 by different lymphocyte subpopulations (CD8+ T cells, CD4+ T cells and B cells). Furthermore, we showed in vivo that pharmacological SIRT1 activation increased the immunological effect of α-PD-1 ICI against melanoma cells in mice, which was accompanied by an increase in T-cell infiltration and T-cell-related cytokines, including interferon (IFN)-γ, CCL4, CXCL9, CXCL10 and tumour necrosis factor-α. In silico analysis of large transcriptional data cohorts showed that SIRT1 was positively associated with the proinflammatory T-cell chemokines CXCL9, CXCL10 and IFN-γ, and prolonged overall survival of patients with melanoma. CONCLUSIONS Our study deciphers the proteomics landscape in human melanoma, providing important information on the tumour microenvironment and identifying SIRT1 as having important prognostic and therapeutic implications.
Collapse
Affiliation(s)
- Jan-Malte Placke
- Department of Dermatology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
- German Cancer Consortium (DKTK), Partner Site Essen/Düsseldorf, Germany
| | - Jenny Bottek
- Institute of Experimental Immunology and Imaging, Department of Immunodynamics, University Hospital Essen, Essen, Germany
| | - Renata Váraljai
- Department of Dermatology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Batool Shannan
- Department of Dermatology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Sarah Scharfenberg
- Department of Dermatology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Christoph Krisp
- Center for Diagnostics, Clinical Chemistry and Laboratory Medicine, University Medical Center Hamburg Eppendorf, Hamburg, Germany
| | - Philippa Spangenberg
- Institute of Experimental Immunology and Imaging, Department of Immunodynamics, University Hospital Essen, Essen, Germany
| | - Camille Soun
- Institute of Experimental Immunology and Imaging, Department of Immunodynamics, University Hospital Essen, Essen, Germany
| | - Devon Siemes
- Institute of Experimental Immunology and Imaging, Department of Immunodynamics, University Hospital Essen, Essen, Germany
| | - Lars Borgards
- Institute of Experimental Immunology and Imaging, Department of Immunodynamics, University Hospital Essen, Essen, Germany
| | - Franziska Hoffmann
- Department of Otorhinolaryngology, Jena University Hospital, Jena, Germany
| | - Fang Zhao
- Department of Dermatology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Anette Paschen
- Department of Dermatology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
- German Cancer Consortium (DKTK), Partner Site Essen/Düsseldorf, Germany
| | - Hartmut Schlueter
- Center for Diagnostics, Clinical Chemistry and Laboratory Medicine, University Medical Center Hamburg Eppendorf, Hamburg, Germany
| | | | - Iris Helfrich
- Department of Dermatology and Allergology, University Hospital, Ludwig Maximilian University, Munich, Germany
| | - Florian Rambow
- German Cancer Consortium (DKTK), Partner Site Essen/Düsseldorf, Germany
- Department of Applied Computational Cancer Research, Institute for AI in Medicine (IKIM), University Hospital Essen, Essen, Germany
| | - Selma Ugurel
- Department of Dermatology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
- German Cancer Consortium (DKTK), Partner Site Essen/Düsseldorf, Germany
| | - Alpaslan Tasdogan
- Department of Dermatology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
- German Cancer Consortium (DKTK), Partner Site Essen/Düsseldorf, Germany
| | - Dirk Schadendorf
- Department of Dermatology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
- German Cancer Consortium (DKTK), Partner Site Essen/Düsseldorf, Germany
| | - Daniel R Engel
- Institute of Experimental Immunology and Imaging, Department of Immunodynamics, University Hospital Essen, Essen, Germany
| | - Alexander Roesch
- Department of Dermatology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
- German Cancer Consortium (DKTK), Partner Site Essen/Düsseldorf, Germany
| |
Collapse
|
5
|
Zhang X, Gao J, Yang L, Feng X, Yuan X. Oxidative stress and its role in recurrent pregnancy loss: mechanisms and implications. J Mol Histol 2024; 56:55. [PMID: 39724438 DOI: 10.1007/s10735-024-10332-z] [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: 09/26/2024] [Accepted: 12/06/2024] [Indexed: 12/28/2024]
Abstract
Recurrent pregnancy loss (RPL) is the occurrence of two or more consecutive miscarriages before 20 weeks of gestation. Recent research has increasingly focused on the role of oxidative stress in RPL, providing insights into its underlying mechanisms and potential therapeutic targets. Oxidative stress arises from an imbalance between reactive oxygen species (ROS) production and antioxidant defenses, leading to cellular damage and inflammation. Oxidative stress has been implicated in disrupting placental blood flow, inducing apoptosis in fetal and placental cells, and exacerbating inflammatory responses, all of which can contribute to pregnancy loss. Elevated levels of ROS have been associated with compromised placental function, impaired fetal development, and increased risk of RPL. Additionally, oxidative stress can modulate maternal immune responses, potentially leading to immune-related pregnancy complications. This review synthesizes current evidence on the mechanisms by which oxidative stress contributes to RPL and highlights emerging research on potential interventions, including antioxidant therapies and lifestyle modifications. Understanding these mechanisms is crucial for developing effective preventive and therapeutic strategies to reduce the risk of RPL and improve pregnancy outcomes. Future research should focus on elucidating the specific pathways involved and exploring novel treatments aimed at mitigating oxidative damage during pregnancy.
Collapse
Affiliation(s)
- Xiaoyu Zhang
- Department of First Clinical Medical College, Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang, 150040, P.R. China
| | - Jiawei Gao
- Department of First Clinical Medical College, Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang, 150040, P.R. China
| | - Liuxin Yang
- Department of First Clinical Medical College, Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang, 150040, P.R. China
| | - Xiaoling Feng
- Department of Gynecology, The First Affiliated Hospital of Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang, 150040, P.R. China.
| | - Xingxing Yuan
- Department of First Clinical Medical College, Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang, 150040, P.R. China.
- Department of Medicine, Heilongjiang Academy of Traditional Chinese Medicine, Harbin, 150006, China.
| |
Collapse
|
6
|
Zhang J, Li N, Hu X. Metabolic Reprograming of Macrophages: A New Direction in Traditional Chinese Medicine for Treating Liver Failure. J Immunol Res 2024; 2024:5891381. [PMID: 39741958 PMCID: PMC11688140 DOI: 10.1155/jimr/5891381] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Revised: 09/03/2024] [Accepted: 11/28/2024] [Indexed: 01/03/2025] Open
Abstract
Acute liver failure (ALF) is a fulminant clinical syndrome that usually leads to multiple organ failure and high mortality. Macrophages play a crucial role in the initiation, development, and recovery of ALF. Targeting macrophages through immunotherapy holds significant promise as a therapeutic strategy. These cells exhibit remarkable plasticity, enabling them to differentiate into various subtypes based on changes in their surrounding microenvironment. M1-type macrophages are associated with a pro-inflammatory phenotype and primarily rely predominantly on glycolysis. In contrast, M2-type macrophages, which are characterized by anti-inflammatory phenotype, predominantly obtain their energy from oxidative phosphorylation (OXPHOS) and fatty acid oxidation (FAO). Shifting macrophage metabolism from glycolysis to OXPHOS inhibits M1 macrophage activation and promotes M2 macrophage activation, thereby exerting anti-inflammatory and reparative effects. This study elucidates the relationship between macrophage activation and glucose metabolism reprograming from an immunometabolism perspective. A comprehensive literature review revealed that several signaling pathways may regulate macrophage polarization through energy metabolism, including phosphatidyl-inositol 3-kinase/protein kinase B (PI3K/AKT), mammalian target of rapamycin (mTOR)/hypoxia-inducible factor 1α (HIF-1α), nuclear factor-κB (NF-κB), and AMP-activated protein kinase (AMPK), which exhibit crosstalk with one another. Additionally, we systematically reviewed several traditional Chinese medicine (TCM) monomers that can modulate glucose metabolism reprograming and influence the polarization states of M1 and M2 macrophages. This review aimed to provide valuable insights that could contribute to the development of new therapies or drugs for ALF.
Collapse
Affiliation(s)
- Junli Zhang
- Department of Infectious Diseases, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
- Department of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Na Li
- Department of Infectious Diseases, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
- Department of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Xiaoyu Hu
- Department of Infectious Diseases, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| |
Collapse
|
7
|
Chen Y, Zhang X, Huang S, Febbraio M. Hidden features: CD36/SR-B2, a master regulator of macrophage phenotype/function through metabolism. Front Immunol 2024; 15:1468957. [PMID: 39742252 PMCID: PMC11685046 DOI: 10.3389/fimmu.2024.1468957] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2024] [Accepted: 11/30/2024] [Indexed: 01/03/2025] Open
Abstract
Once thought to be in a terminally differentiated state, macrophages are now understood to be highly pliable, attuned and receptive to environmental cues that control and align responses. In development of purpose, the centrality of metabolic pathways has emerged. Thus, macrophage inflammatory or reparative phenotypes are tightly linked to catabolic and anabolic metabolism, with further fine tuning of specific gene expression patterns in specific settings. Single-cell transcriptome analyses have revealed a breadth of macrophage signatures, with some new influencers driving phenotype. CD36/Scavenger Receptor B2 has established roles in immunity and lipid metabolism. Macrophage CD36 is a key functional player in metabolic expression profiles that determine phenotype. Emerging data show that alterations in the microenvironment can recast metabolic pathways and modulate macrophage function, with the potential to be leveraged for therapeutic means. This review covers recent data on phenotypic characterization of homeostatic, atherosclerotic, lipid-, tumor- and metastatic-associated macrophages, with the integral role of CD36 highlighted.
Collapse
Affiliation(s)
- Yuge Chen
- Mike Petryk School of Dentistry, Faculty of Medicine and Dentistry, College of Health Sciences, University of Alberta, Edmonton, AB, Canada
| | - Xuejia Zhang
- Mike Petryk School of Dentistry, Faculty of Medicine and Dentistry, College of Health Sciences, University of Alberta, Edmonton, AB, Canada
- School and Hospital of Stomatology, Wenzhou Medical University, Wenzhou, China
| | - Shengbin Huang
- Institute of Stomatology, School and Hospital of Stomatology, Wenzhou Medical University, Wenzhou, China
- Department of Prosthodontics, School and Hospital of Stomatology, Wenzhou Medical University, Wenzhou, China
| | - Maria Febbraio
- Mike Petryk School of Dentistry, Faculty of Medicine and Dentistry, College of Health Sciences, University of Alberta, Edmonton, AB, Canada
| |
Collapse
|
8
|
Wang S, Sun T, Huang Y, Mi R, Zhang Y, Gong H, Chen Z. Levonorgestrel enhanced Toxoplasma gondii infection risk via progesterone receptor upregulation. Vet Parasitol 2024; 332:110330. [PMID: 39427455 DOI: 10.1016/j.vetpar.2024.110330] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Revised: 09/14/2024] [Accepted: 10/09/2024] [Indexed: 10/22/2024]
Abstract
The numerous stray cats and dogs worldwide pose a huge burden on local governments, keeping residents safe and healthy, and maintaining clean cities with good traffic situations. To effectively control the populations of these stray animals, while considering animal welfare, researchers have suggested the use of levonorgestrel (LNG) as a contraceptive method. However, the potential side effects of LNG on these animals need to be evaluated before widespread application. Since dogs and cats play important roles in the transmission of Toxoplasma gondii as intermediate and definitive hosts, respectively, it is critical to assess the safety of LNG from the perspective of its effect on Toxoplasma susceptibility. In this study, the susceptibility of mice to T. gondii infection was investigated in vivo and in vitro with the presence of LNG. As a result, LNG increased the risk of infection, which was probably associated with the downregulation of IFN-γ levels, but not with the alteration of host sex hormone levels. Concurrently, the presence of LNG enhanced the expression of its ligand, the progesterone receptor (PGR), on the host cells. The promotional effect of LNG on T. gondii infection was attenuated when the PGR gene was knocked down. This investigation represents the inaugural study of LNG's side effects on T. gondii infection in mice, underscoring the significance of choosing or developing suitable contraceptive drugs for stray animals.
Collapse
Affiliation(s)
- Shenghui Wang
- Key Laboratory of Animal Parasitology of Ministry of Agriculture and Rural Affairs, Laboratory of Quality and Safety Risk Assessment for Animal Products on Biohazards (Shanghai) of Ministry of Agriculture and Rural Affairs, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai 200241, China
| | - Tiancong Sun
- Key Laboratory of Animal Parasitology of Ministry of Agriculture and Rural Affairs, Laboratory of Quality and Safety Risk Assessment for Animal Products on Biohazards (Shanghai) of Ministry of Agriculture and Rural Affairs, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai 200241, China
| | - Yan Huang
- Key Laboratory of Animal Parasitology of Ministry of Agriculture and Rural Affairs, Laboratory of Quality and Safety Risk Assessment for Animal Products on Biohazards (Shanghai) of Ministry of Agriculture and Rural Affairs, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai 200241, China
| | - Rongsheng Mi
- Key Laboratory of Animal Parasitology of Ministry of Agriculture and Rural Affairs, Laboratory of Quality and Safety Risk Assessment for Animal Products on Biohazards (Shanghai) of Ministry of Agriculture and Rural Affairs, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai 200241, China
| | - Yan Zhang
- Key Laboratory of Animal Parasitology of Ministry of Agriculture and Rural Affairs, Laboratory of Quality and Safety Risk Assessment for Animal Products on Biohazards (Shanghai) of Ministry of Agriculture and Rural Affairs, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai 200241, China
| | - Haiyan Gong
- Key Laboratory of Animal Parasitology of Ministry of Agriculture and Rural Affairs, Laboratory of Quality and Safety Risk Assessment for Animal Products on Biohazards (Shanghai) of Ministry of Agriculture and Rural Affairs, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai 200241, China.
| | - Zhaoguo Chen
- Key Laboratory of Animal Parasitology of Ministry of Agriculture and Rural Affairs, Laboratory of Quality and Safety Risk Assessment for Animal Products on Biohazards (Shanghai) of Ministry of Agriculture and Rural Affairs, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai 200241, China.
| |
Collapse
|
9
|
Liu JJ, Yang JB, Wang Y, Hu XR, Wang YD, Nie LX, Wei F, Yu JD, Yao LW, Xu BL, Ma SC, Jin HY. Integrating network pharmacology and experimental validation to investigate the effects and mechanism of Renshen Shouwu decoction for ameliorating Alzheimer's disease. PHARMACEUTICAL BIOLOGY 2024; 62:767-780. [PMID: 39417324 PMCID: PMC11488172 DOI: 10.1080/13880209.2024.2415660] [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: 12/04/2023] [Revised: 09/29/2024] [Accepted: 10/08/2024] [Indexed: 10/19/2024]
Abstract
CONTEXT The mechanism of Renshen Shouwu Decoction (RSSW) in treating Alzheimer's disease (AD) remains unknown. OBJECTIVE This study investigates the effects and mechanism of RSSW for ameliorating AD. MATERIALS AND METHODS Ten SAMR1 mice and 40 SAMP8 mice were divided into five groups: control (SAMR1), model (SAMP8), positive drug (Donepezil, 1.3 mg/kg/d), and RSSW (Low-dose, 117 mg/kg/d; High-dose, 234 mg/kg/d). Starting from 6 months of age, the medications were administered intragastrically for a total of 60 days. Subsequently, memory improvement in rapidly aging mice was assessed using the novel object recognition test and Morris water maze test. Through the identification of absorbed blood components and analysis of network pharmacology, active ingredients and potential targets involved in the treatment of AD were identified. Finally, AD-related biological indicators were detected using western blotting and ELISA. RESULT Our results demonstrated that RSSW effectively ameliorated memory impairments, inhibited tau hyperphosphorylation, and reduced β-amyloid plaque deposition in SAMP8 mice. Thirty absorbed blood components in RSSW were identified, revealing identified 96 major targets that play a key role in alleviating AD. Notably, the obtained main targets were highly enriched in SIRT1-mediated signaling pathways. Subsequent experimental validation confirmed that RSSW activated the SIRT1/NF-κB, SIRT1/AMPK, and SIRT1/p53 signaling cascades. Nine potential active ingredients were predicted through molecular docking. DISCUSSION AND CONCLUSIONS Our research findings suggest the mechanism of RSSW treatment for AD, which ameliorates memory impairments by reducing cortical tissue inflammation and apoptosis.
Collapse
Affiliation(s)
- Jing-jing Liu
- Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
- National Institutes for Food and Drug Control, Beijing, China
| | - Jian-bo Yang
- National Institutes for Food and Drug Control, Beijing, China
| | - Ying Wang
- National Institutes for Food and Drug Control, Beijing, China
| | - Xiao-ru Hu
- National Institutes for Food and Drug Control, Beijing, China
| | - Ya-dan Wang
- National Institutes for Food and Drug Control, Beijing, China
| | - Li-xing Nie
- National Institutes for Food and Drug Control, Beijing, China
| | - Feng Wei
- National Institutes for Food and Drug Control, Beijing, China
| | - Jian-dong Yu
- National Institutes for Food and Drug Control, Beijing, China
| | - Ling-wen Yao
- National Institutes for Food and Drug Control, Beijing, China
| | - Bei-lei Xu
- School of Pharmacy, Harbin University of Commerce, Harbin, China
| | - Shuang-cheng Ma
- Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
- National Institutes for Food and Drug Control, Beijing, China
| | - Hong-yu Jin
- Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| |
Collapse
|
10
|
Sun H, Li D, Wei C, Liu L, Xin Z, Gao H, Gao R. The relationship between SIRT1 and inflammation: a systematic review and meta-analysis. Front Immunol 2024; 15:1465849. [PMID: 39676853 PMCID: PMC11638041 DOI: 10.3389/fimmu.2024.1465849] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Accepted: 11/07/2024] [Indexed: 12/17/2024] Open
Abstract
Recent studies underscore the anti-inflammatory role of SIRT1; however, its levels during inflammatory states remain ambiguous. We synthesized relevant studies up to 20 March 2024 to evaluate the relationship between SIRT1 and inflammation, using data from three major databases. Employing a random-effects model, we analyzed both cross-sectional and longitudinal studies, calculating weighted mean differences (WMDs) for pooled effect sizes. Subgroup and sensitivity analyses, along with a risk of bias assessment, were also conducted. We reviewed 13 publications, encompassing 21 datasets and 2,028 participants. The meta-analysis indicated higher SIRT1 levels in inflammatory groups compared to control groups pre-adjustment (WMD, 3.18 ng/ml; 95% CI 2.30, 4.06 ng/ml; P<0.001; I²= 99.7%) and post-adjustment (WMD, 0.88 ng/ml; 95% CI 0.14, 1.62 ng/ml; P<0.001; I²= 99.5%). Notably, middle-aged patients with inflammation exhibited lower SIRT1 levels (WMD, -0.85 ng/ml; 95% CI -1.47, -0.22 ng/ml; P= 0.008; I²= 95.4%), while groups characterized by East Asian descent, plasma studies, autoimmune conditions, and musculoskeletal disorders showed higher levels. The findings suggest that inflammation generally upregulates SIRT1, potentially elucidating its role in immunobiological processes. However, the significant heterogeneity observed, partly due to the cross-sectional nature of some data, limits insights into the duration of disease progression, which remains highly variable.
Collapse
Affiliation(s)
- Haiyang Sun
- Department of Immunology, College of Basic Medical Sciences, Jilin University, Changchun, China
| | - Dong Li
- Department of Immunology, College of Basic Medical Sciences, Jilin University, Changchun, China
| | - Chaojie Wei
- Department of Immunology, College of Basic Medical Sciences, Jilin University, Changchun, China
| | - Liping Liu
- Department of Immunology, College of Basic Medical Sciences, Jilin University, Changchun, China
| | - Zhuoyuan Xin
- Key Laboratory of Zoonosis Research, Ministry of Education, College of Basic Medical Science, Jilin University, Changchun, China
| | - Hang Gao
- Department of Bone and Joint Surgery, Orthopaedic Surgery Center, The First Hospital of Jilin University, Changchun, China
| | - Rong Gao
- Department of Respiratory and Critical Care Medicine, The Second Hospital of Jilin University, Changchun, China
| |
Collapse
|
11
|
Fernandez F, Griffiths LR, Sutherland HG, Cole MH, Fitton JH, Winberg P, Schweitzer D, Hopkins LN, Meyer BJ. Sirtuin Proteins and Memory: A Promising Target in Alzheimer's Disease Therapy? Nutrients 2024; 16:4088. [PMID: 39683482 DOI: 10.3390/nu16234088] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2024] [Revised: 11/20/2024] [Accepted: 11/25/2024] [Indexed: 12/18/2024] Open
Abstract
Sirtuins (SIRTs), nicotine adenine dinucleotide (+)-dependent histone deacetylases, have emerged as critical regulators in many signalling pathways involved in a wide range of biological processes. Currently, seven mammalian SIRTs have been characterized and are found across a number of cellular compartments. There has been considerable interest in the role of SIRTs in the brain due to their role in a plethora of metabolic- and age-related diseases, including their involvement in learning and memory function in physiological and pathophysiological conditions. Although cognitive function declines over the course of healthy ageing, neurological disorders including Alzheimer's disease (AD) can be associated with progressive cognitive impairments. This review aimed to report and integrate recent advances in the understanding of the role of SIRTs in cognitive function and dysfunction in the context of AD. We have also reviewed the use of selective and/or natural SIRT activators as potential therapeutic agents and/or adjuvants for AD.
Collapse
Affiliation(s)
- Francesca Fernandez
- School of Behavioural and Health Sciences, Faculty of Heath Sciences, Australian Catholic University, Banyo, QLD 4014, Australia
- Centre for Genomics and Personalised Health, School of Biomedical Sciences, Queensland University of Technology, 60 Musk Ave, Kelvin Grove, QLD 4059, Australia
- Healthy Brain and Mind Research Centre, Australian Catholic University, Fitzroy, VIC 3065, Australia
| | - Lyn R Griffiths
- Centre for Genomics and Personalised Health, School of Biomedical Sciences, Queensland University of Technology, 60 Musk Ave, Kelvin Grove, QLD 4059, Australia
| | - Heidi G Sutherland
- Centre for Genomics and Personalised Health, School of Biomedical Sciences, Queensland University of Technology, 60 Musk Ave, Kelvin Grove, QLD 4059, Australia
| | - Michael H Cole
- School of Behavioural and Health Sciences, Faculty of Heath Sciences, Australian Catholic University, Banyo, QLD 4014, Australia
- Healthy Brain and Mind Research Centre, Australian Catholic University, Fitzroy, VIC 3065, Australia
| | - J Helen Fitton
- Venus Shell Systems Pty Ltd., Huskisson, NSW 2540, Australia
| | - Pia Winberg
- Venus Shell Systems Pty Ltd., Huskisson, NSW 2540, Australia
- School of Medical, Indigenous and Health Science, University of Wollongong, Wollongong, NSW 2522, Australia
| | - Daniel Schweitzer
- Mater Centre of Neuroscience, 53 Raymond Terrace, South Brisbane, QLD 4066, Australia
- Department of Neurology, Wesley Hospital, 451 Coronation Drive, Auchenflower, QLD 4066, Australia
| | - Lloyd N Hopkins
- Centre for Genomics and Personalised Health, School of Biomedical Sciences, Queensland University of Technology, 60 Musk Ave, Kelvin Grove, QLD 4059, Australia
| | - Barbara J Meyer
- School of Medical, Indigenous and Health Science, University of Wollongong, Wollongong, NSW 2522, Australia
- Molecular Horizons, University of Wollongong, Wollongong, NSW 2522, Australia
| |
Collapse
|
12
|
Wen B, Huang Y, Deng G, Yan Q, Jia L. Gut microbiota analysis and LC-MS-based metabolomics to investigate AMPK/NF-κB regulated by Clostridium butyricum in the treatment of acute pancreatitis. J Transl Med 2024; 22:1072. [PMID: 39604956 PMCID: PMC11600808 DOI: 10.1186/s12967-024-05764-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Accepted: 10/14/2024] [Indexed: 11/29/2024] Open
Abstract
BACKGROUND Acute pancreatitis (AP) is an inflammatory condition with potentially life-threatening complications. This study investigates the therapeutic potential of Clostridium butyricum for modulating the inflammatory cascade through the AMPK/NF-κB signaling pathway, focusing on inflammation induced by AP. LC-MS analysis of serum samples from AP patients highlighted the regulation of lipid metabolism and inflammation, and found that metabolites involved in the inhibition of NF-κB phosphorylation and the AMPK activation pathway were downregulated. We hypothesized that pre-administration of Clostridium butyricum and its culture supernatant could mitigate AP-induced damage by modulating the AMPK/NF-κB pathway. METHODS Lipopolysaccharide (LPS)-induced cell inflammation models. LPS combined with CAE induced acute pancreatitis in mice. We divided mice into four groups: Con, AP, AP + C.Buty (AP with Clostridium butyricum treatment), and AP + CFS (AP with culture supernatant treatment). Analyses were performed using WB, RT-qPCR, Elisa, flow cytometry, IHC, and HE, respectively. RESULTS Our study shows that CFS can reduce the apoptosis of LPS-induced cellular inflammation and reduce the release of LPS-induced cytoinflammatory factors through the AMPK/NF-κB pathway in vitro. In vivo, Clostridium butyricum and its supernatant significantly reduced inflammatory markers, and corrected histopathological alterations in AP mice. Gut microbiota analysis further supported these results, showing that Clostridium butyricum and its supernatant could restore the balance of intestinal flora disrupted by AP. CONCLUSIONS Mechanistically, our results indicated that the therapeutic effects of Clostridium butyricum are mediated through the activation of AMPK, leading to the inhibition of the NF-κB pathway, thereby reducing the production of pro-inflammatory cytokines. Clostridium butyricum and its culture supernatant exert a protective effect against AP-induced damage by modulating the AMPK/NF-κB signaling pathway. Future studies will further elucidate the molecular mechanisms underlying the beneficial effects of Clostridium butyricum in AP and explore its clinical applicability in human subjects.
Collapse
Affiliation(s)
- Biyan Wen
- Department of Gastroenterology, The Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, 510013, China
- Department of Gastroenterology, Guangzhou First People's Hospital, South China University of Technology, Guangzhou, 510180, China
| | - Yaoxing Huang
- Department of Gastroenterology, Guangzhou First People's Hospital, South China University of Technology, Guangzhou, 510180, China
| | - Guiqing Deng
- Department of Gastroenterology, The Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, 510013, China
| | - Qingqing Yan
- Department of Gastroenterology, Guangzhou First People's Hospital, South China University of Technology, Guangzhou, 510180, China
| | - Lin Jia
- Department of Gastroenterology, The Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, 510013, China.
- Department of Gastroenterology, Guangzhou First People's Hospital, South China University of Technology, Guangzhou, 510180, China.
| |
Collapse
|
13
|
Aziz A, Ganesan Nathan K, Kamarul T, Mobasheri A, Sharifi A. The interplay between dysregulated metabolites and signaling pathway alterations involved in osteoarthritis: a systematic review. Ther Adv Musculoskelet Dis 2024; 16:1759720X241299535. [PMID: 39600593 PMCID: PMC11590150 DOI: 10.1177/1759720x241299535] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2024] [Accepted: 10/24/2024] [Indexed: 11/29/2024] Open
Abstract
Background Osteoarthritis (OA) is a common degenerative joint disease that poses a significant global healthcare challenge due to its complexity and limited treatment options. Advances in metabolomics have provided insights into OA by identifying dysregulated metabolites and their connection to altered signaling pathways. However, a comprehensive understanding of these biomarkers in OA is still required. Objectives This systematic review aims to identify metabolomics biomarkers associated with dysregulated signaling pathways in OA, using data from various biological samples, including in vitro models, animal studies, and human research. Design A systematic review was conducted following Preferred Reporting Items for Systematic Reviews and Meta-Analyses guidelines. Data sources and methods Data were gathered from literature published between August 2017 and May 2024, using databases such as "PubMed," "Scopus," "Web of Science," and "Google Scholar." Studies were selected based on keywords like "metabolomics," "osteoarthritis," "amino acids," "molecular markers," "biomarkers," "diagnostic markers," "inflammatory cytokines," "molecular signaling," and "signal transduction." The review focused on identifying key metabolites and their roles in OA-related pathways. Limitations include the potential exclusion of studies due to keyword selection and strict inclusion criteria. Results The meta-analysis identified dysregulated metabolites and associated pathways, highlighting a distinct set of related metabolites consistently altered across the studies analyzed. The dysregulated metabolites, including amino acids, lipids, and carbohydrates, were found to play critical roles in inflammation, oxidative stress, and energy metabolism in OA. Metabolites such as alanine, lysine, and proline were frequently linked to pathways involved in inflammation, cartilage degradation, and apoptosis. Key pathways, including nuclear factor kappa B, mitogen-activated protein kinase, Wnt/β-catenin, and mammalian target of rapamycin, were associated with changes in metabolite levels, particularly in proinflammatory lipids and energy-related compounds. Conclusion This review reveals a complex interplay between dysregulated metabolites and signaling pathways in OA, offering potential biomarkers and therapeutic targets. Further research is needed to explore the molecular mechanisms driving these changes and their implications for OA treatment.
Collapse
Affiliation(s)
- Atiqah Aziz
- Tissue Engineering Group, National Orthopaedic Centre of Excellence for Research and Learning, Department of Orthopaedic Surgery, Faculty of Medicine, Universiti Malaya, Kuala Lumpur, Malaysia
| | - Kavitha Ganesan Nathan
- Tissue Engineering Group, National Orthopaedic Centre of Excellence for Research and Learning, Department of Orthopaedic Surgery, Faculty of Medicine, Universiti Malaya, Kuala Lumpur, Malaysia
| | - Tunku Kamarul
- Tissue Engineering Group, National Orthopaedic Centre of Excellence for Research and Learning, Department of Orthopaedic Surgery, Faculty of Medicine, Universiti Malaya, Kuala Lumpur, Malaysia
| | - Ali Mobasheri
- Research Unit of Health Sciences and Technology, Faculty of Medicine, University of Oulu, Oulu, Finland
- Department of Regenerative Medicine, State Research Institute Centre for Innovative Medicine, Vilnius, Lithuania
- Department of Joint Surgery, First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
- World Health Organization Collaborating Centre for Public Health Aspects of Musculoskeletal Health and Aging, Université de Liège, Liège, Belgium
| | - Alimohammad Sharifi
- Tissue Engineering Group, National Orthopaedic Centre of Excellence for Research and Learning, Department of Orthopaedic Surgery, Faculty of Medicine, University of Malaya, Kuala Lumpur 50603, Malaysia
- Department of Pharmacology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
- Stem cell and Regenerative Medicine Research Center, Iran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
14
|
Taghizadeh M, Maleki MH, Vakili O, Tavakoli R, Zarei P, Dehghanian A, Bordbar H, Shafiee SM. Bilirubin, a hepatoprotective agent that activates SIRT1, PGC-1α, and PPAR-α, while inhibiting NF-κB in rats with metabolic-associated fatty liver disease. Sci Rep 2024; 14:29244. [PMID: 39587213 PMCID: PMC11589846 DOI: 10.1038/s41598-024-80119-5] [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: 08/05/2024] [Accepted: 11/15/2024] [Indexed: 11/27/2024] Open
Abstract
Metabolic-associated fatty liver disease (MAFLD) is a chronic liver disorder characterized by fatty liver disease alongside overweight or obesity and/or type 2 diabetes mellitus (T2DM). Timely intervention is crucial for a potential cure. This study aimed to investigate the effects of bilirubin, an endogenous antioxidant, on lipid metabolism and inflammation in MAFLD. Specifically, it examined bilirubin's impact on SIRT1, PPAR-α, and NF-κB in the livers of rats with MAFLD induced by a high-fat diet (HFD) and streptozotocin (STZ) administration. Forty eight-week adult male Sprague Dawley rats were divided into five groups (n = 8): Control, HFD-STZ, HFD-S-BR6, HFD-S-BR14, and C-BR14. In the last three groups, bilirubin administration was performed intraperitoneally for 6 and 14 weeks (10 mg/kg/day). We selected the key genes associated with MAFLD and subsequently performed GO (Gene Ontology) and KEGG (Kyoto Encyclopedia of Genes and Genomes) analyses to explore the enriched biological processes and signaling pathways. Hence, the gene expression of SIRT1, PGC-1α, PPAR-α, and inflammatory genes (NF-κB, TNF-α, IL-6, and IL-1β) was measured using Real-time quantitative PCR. Stereological and histopathological alterations of liver structure as well as lipid profile, biochemical indices, and liver indices, were also assessed among different groups. The enrichment analysis identified that several signaling pathways and biological processes might be related to MAFLD. Bilirubin-treated rats contained higher PPAR-α, PGC-1α, and SIRT1 expression levels by approximately 5.7-, 2.1-, and 2.2-fold, respectively, compared to the HFD-receiving rats (p < 0.0001, p < 0.05, and p < 0.05). Whereas, the genes involved in the inflammatory cascades, including NF-κB, TNF-α, and IL-6, were downregulated by 0.6-fold (p < 0.05) following 14-week treatment of bilirubin, while only significantly decreased expression of NF-κB and IL-6 (approximately 0.6-fold, p < 0.05) were observed after 6-week treatment of bilirubin. Remarkably, bilirubin administration favorably reversed the effects of HFD on the liver's volume and cell numbers and ameliorated the related structural changes. It also improved lipid profile, biochemical parameters, and liver indices of HFD-STZ rats. This study indicated that bilirubin acts as a protective/ameliorative compound against MAFLD, particularly through regulating the key genes involved in lipid metabolism and inflammation in HFD-STZ rats.
Collapse
Affiliation(s)
- Motahareh Taghizadeh
- Student Research Committee, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
- Department of Clinical Biochemistry, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mohammad Hasan Maleki
- Department of Clinical Biochemistry, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Omid Vakili
- Department of Clinical Biochemistry, School of Pharmacy and Pharmaceutical Sciences, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Ramin Tavakoli
- Student Research Committee, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
- Department of Clinical Biochemistry, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Parvin Zarei
- Department of Bioinformatics, School of Advanced Technologies in Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Amirreza Dehghanian
- Trauma Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
- Molecular Pathology and Cytogenetics Division, Department of Pathology, School of Medicine, Shiraz University, Shiraz, Iran
| | - Hossein Bordbar
- Histomorphometry and Stereology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
- Department of Anatomy, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Sayed Mohammad Shafiee
- Autophagy Research Center, Department of Clinical Biochemistry, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran.
| |
Collapse
|
15
|
Wang X, Song X, Fang K, Chang X. CD38 modulates cytokine secretion by NK cells through the Sirt1/NF-κB pathway, suppressing immune surveillance in colorectal cancer. Sci Rep 2024; 14:28702. [PMID: 39562615 PMCID: PMC11577103 DOI: 10.1038/s41598-024-79008-8] [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: 01/12/2024] [Accepted: 11/05/2024] [Indexed: 11/21/2024] Open
Abstract
Tregs and M2-type macrophages are essential for immune surveillance. CD38 + NK cells are involved in immunoregulation by modulating cytokine secretion. This study investigated how CD38 + NKs affect Tregs and macrophages in colorectal cancer (CRC). Higher proportions of CD38 + NKs and Tregs were detected in bloods and tumor tissues of CRC patients than that in the samples from healthy controls (HCs). Compared with CD38 + NKs from HCs, the NK cells from CRC promoted the differentiation of Tregs from CD4 + T cells, and secreted increased levels of IL-10, TGF-β and TNF-α and decreased levels of IFN-γ. CD38 + NKs from CRC expressed higher levels of CD38, NF-κB and acetyl-NF-κB and lower levels of Sirt1. When CRC CD38 + NK cells were treated with anti-CD38 monoclonal antibody, the above trends were reversed. CRC CD38 + NKs with treatment of NF-κB inhibitor also showed opposite effects on cytokine secretion and CD4 + T-cell differentiation. After treatment with a Sirt1 activator, NF-κB signaling was inhibited in these CD38 + NKs, whereas treatment with a Sirt1 inhibitor activated NF-κB signaling. The supernatants of CRC CD38 + NK culture promoted M0 macrophage polarization to M2-type. We suggest that CD38 modulates cytokine secretion by NK cells through Sirt1/NF-κB signaling pathway, thereby suppressing immune surveillance in tumorigenesis.
Collapse
Affiliation(s)
- Xueling Wang
- Medical Research Center, The Affiliated Hospital of Qingdao University, Wutaishan road 1677, Qingdao, 266000, People's Republic of China
- Qingdao Municipal Hospital, University of Health and Rehabilitation Sciences, Jiaozhou road 1, Qingdao, 266000, People's Republic of China
| | - Xianqin Song
- Medical Research Center, The Affiliated Hospital of Qingdao University, Wutaishan road 1677, Qingdao, 266000, People's Republic of China
| | - Kehua Fang
- Clinical Laboratory, The Affiliated Hospital of Qingdao University, Wutaishan road 1677, Qingdao, 266000, Shandong, People's Republic of China.
| | - Xiaotian Chang
- Medical Research Center, The Affiliated Hospital of Qingdao University, Wutaishan road 1677, Qingdao, 266000, People's Republic of China.
| |
Collapse
|
16
|
Cendrowski J, Wrobel M, Mazur M, Jary B, Maurya R, Wang S, Korostynski M, Dziewulska A, Rohm M, Kuropka P, Pudelko-Malik N, Mlynarz P, Dobrzyn A, Zeigerer A, Miaczynska M. NFκB and JNK pathways mediate metabolic adaptation upon ESCRT-I deficiency. Cell Mol Life Sci 2024; 81:458. [PMID: 39560723 DOI: 10.1007/s00018-024-05490-y] [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: 03/25/2024] [Revised: 09/17/2024] [Accepted: 10/24/2024] [Indexed: 11/20/2024]
Abstract
Endosomal Sorting Complexes Required for Transport (ESCRTs) are crucial for delivering membrane receptors or intracellular organelles for lysosomal degradation which provides the cell with lysosome-derived nutrients. Yet, how ESCRT dysfunction affects cell metabolism remained elusive. To address this, we analyzed transcriptomes of cells lacking TSG101 or VPS28 proteins, components of ESCRT-I subcomplex. ESCRT-I deficiency reduced the expression of genes encoding enzymes involved in oxidation of fatty acids and amino acids, such as branched-chain amino acids, and increased the expression of genes encoding glycolytic enzymes. The changes in metabolic gene expression were associated with Warburg effect-like metabolic reprogramming that included intracellular accumulation of lipids, increased glucose/glutamine consumption and lactate production. Moreover, depletion of ESCRT-I components led to expansion of the ER and accumulation of small mitochondria, most of which retained proper potential and performed ATP-linked respiration. Mechanistically, the observed transcriptional reprogramming towards glycolysis in the absence of ESCRT-I occurred due to activation of the canonical NFκB and JNK signaling pathways and at least in part by perturbed lysosomal degradation. We propose that by activating the stress signaling pathways ESCRT-I deficiency leads to preferential usage of extracellular nutrients, like glucose and glutamine, for energy production instead of lysosome-derived nutrients, such as fatty acids and branched-chain amino acids.
Collapse
Affiliation(s)
- Jaroslaw Cendrowski
- Laboratory of Cell Biology, International Institute of Molecular and Cell Biology, Warsaw, Poland.
- Department of Genetics, Maria Sklodowska-Curie National Research Institute of Oncology, Warsaw, Poland.
| | - Marta Wrobel
- Laboratory of Cell Biology, International Institute of Molecular and Cell Biology, Warsaw, Poland
| | - Michal Mazur
- Laboratory of Cell Biology, International Institute of Molecular and Cell Biology, Warsaw, Poland
| | - Bartosz Jary
- Laboratory of Cell Biology, International Institute of Molecular and Cell Biology, Warsaw, Poland
| | - Ranjana Maurya
- Laboratory of Cell Biology, International Institute of Molecular and Cell Biology, Warsaw, Poland
| | - Surui Wang
- Institute for Diabetes and Cancer, Helmholtz Munich, Neuherberg, Germany
- Joint Heidelberg-IDC Translational Diabetes Program, Inner Medicine 1, University Hospital, Heidelberg, Germany
- German Center for Diabetes Research, Neuherberg, Germany
| | - Michal Korostynski
- Laboratory of Pharmacogenomics, Department of Molecular Neuropharmacology, Institute of Pharmacology Polish Academy of Sciences, Krakow, Poland
| | - Anna Dziewulska
- Laboratory of Cell Signaling and Metabolic Disorders, Nencki Institute of Experimental Biology, Warsaw, Poland
| | - Maria Rohm
- Institute for Diabetes and Cancer, Helmholtz Munich, Neuherberg, Germany
- Joint Heidelberg-IDC Translational Diabetes Program, Inner Medicine 1, University Hospital, Heidelberg, Germany
- German Center for Diabetes Research, Neuherberg, Germany
| | - Patryk Kuropka
- Department of Biochemistry, Molecular Biology and Biotechnology, Faculty of Chemistry, Wroclaw University of Science and Technology, Wroclaw, Poland
| | - Natalia Pudelko-Malik
- Department of Biochemistry, Molecular Biology and Biotechnology, Faculty of Chemistry, Wroclaw University of Science and Technology, Wroclaw, Poland
| | - Piotr Mlynarz
- Department of Biochemistry, Molecular Biology and Biotechnology, Faculty of Chemistry, Wroclaw University of Science and Technology, Wroclaw, Poland
| | - Agnieszka Dobrzyn
- Laboratory of Cell Signaling and Metabolic Disorders, Nencki Institute of Experimental Biology, Warsaw, Poland
| | - Anja Zeigerer
- Institute for Diabetes and Cancer, Helmholtz Munich, Neuherberg, Germany
- Joint Heidelberg-IDC Translational Diabetes Program, Inner Medicine 1, University Hospital, Heidelberg, Germany
- German Center for Diabetes Research, Neuherberg, Germany
- European Center for Angioscience (ECAS), Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Marta Miaczynska
- Laboratory of Cell Biology, International Institute of Molecular and Cell Biology, Warsaw, Poland.
| |
Collapse
|
17
|
Deng L, Xie W, Lin M, Xiong D, Huang L, Zhang X, Qian R, Huang X, Tang S, Liu W. Taraxerone inhibits M1 polarization and alleviates sepsis-induced acute lung injury by activating SIRT1. Chin Med 2024; 19:159. [PMID: 39543653 PMCID: PMC11566926 DOI: 10.1186/s13020-024-01002-z] [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: 04/22/2024] [Accepted: 09/08/2024] [Indexed: 11/17/2024] Open
Abstract
BACKGROUND Acute lung injury (ALI) is the most lethal disease associated with sepsis, and there is a lack of effective drug treatment. As the major cells of sepsis-induced ALI, macrophages polarize toward the proinflammatory M1 phenotype and secrete multiple inflammatory cytokines to accelerate the disease process through nuclear factor kappa-B (NF-κB) and NLR family pyrin domain containing 3 (NLRP3) inflammasome signaling pathways. Taraxerone, the main component of the Chinese medicinal Sedum, possesses numerous biological activities. However, uncertainty remains regarding the potential of taraxerone to protect against sepsis-induced ALI. This study aimed to investigate the effects and mechanisms of taraxerone against ALI. METHODS An animal model for ALI was established by cecal ligation and puncture and treated with taraxerone via intraperitoneal administration. The protective effect of taraxerone on the lungs was analyzed using H&E staining, dihydroethidium staining, ELISA kits, cell counting, myeloperoxidase kit, malondialdehyde kit, glutathione kit, superoxide dismutase kit and flow cytometry. Western blotting, RT-PCR, flow cytometry, co-immunoprecipitation, and immunofluorescence were used to investigate the regulatory of taraxerone on SIRT1. RESULTS Our study demonstrates for the first time that taraxerone can activate SIRT1 in macrophages, promoting SIRT1 activity. This activation inhibited the NF-κB signaling pathway primarily through the dephosphorylation and deacetylation of p65. Simultaneously, taraxerone disrupted the NLRP3 inflammasome signaling pathway, thereby alleviating M1 polarization of macrophages and mitigating sepsis-induced pulmonary inflammation and oxidative stress. In vivo, EX527 was used to validate the anti-inflammatory and anti-oxidative stress effects of taraxerone mediated by SIRT1. CONCLUSION SIRT1-mediated anti-inflammatory and anti-oxidative stress effects may be important targets for taraxerone in treating ALI.
Collapse
Affiliation(s)
- Lang Deng
- Xiangya Nursing School, Central South University, Changsha, 410013, Hunan, China
| | - Weixi Xie
- Xiangya Nursing School, Central South University, Changsha, 410013, Hunan, China
| | - Miao Lin
- Xiangya Nursing School, Central South University, Changsha, 410013, Hunan, China
| | - Dayan Xiong
- Xiangya Nursing School, Central South University, Changsha, 410013, Hunan, China
| | - Lei Huang
- Occupational Disease Department, Hunan Prevention and Treatment Institute for Occupational Diseases, Changsha, 410021, Hunan, China
| | - Xiaohua Zhang
- Occupational Disease Department, Hunan Prevention and Treatment Institute for Occupational Diseases, Changsha, 410021, Hunan, China
| | - Rui Qian
- Xiangya Nursing School, Central South University, Changsha, 410013, Hunan, China
| | - Xiaoting Huang
- Xiangya Nursing School, Central South University, Changsha, 410013, Hunan, China.
| | - Siyuan Tang
- Xiangya Nursing School, Central South University, Changsha, 410013, Hunan, China.
| | - Wei Liu
- Xiangya Nursing School, Central South University, Changsha, 410013, Hunan, China.
| |
Collapse
|
18
|
Yu W, Yu Y, Sun S, Lu C, Zhai J, Lei Y, Bai F, Wang R, Chen J. Immune Alterations with Aging: Mechanisms and Intervention Strategies. Nutrients 2024; 16:3830. [PMID: 39599617 PMCID: PMC11597283 DOI: 10.3390/nu16223830] [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: 09/28/2024] [Revised: 10/31/2024] [Accepted: 11/06/2024] [Indexed: 11/29/2024] Open
Abstract
Aging is the result of a complex interplay of physical, environmental, and social factors, leading to an increased prevalence of chronic age-related diseases that burden health and social care systems. As the global population ages, it is crucial to understand the aged immune system, which undergoes declines in both innate and adaptive immunity. This immune decline exacerbates the aging process, creating a feedback loop that accelerates the onset of diseases, including infectious diseases, autoimmune disorders, and cancer. Intervention strategies, including dietary adjustments, pharmacological treatments, and immunomodulatory therapies, represent promising approaches to counteract immunosenescence. These interventions aim to enhance immune function by improving the activity and interactions of aging-affected immune cells, or by modulating inflammatory responses through the suppression of excessive cytokine secretion and inflammatory pathway activation. Such strategies have the potential to restore immune homeostasis and mitigate age-related inflammation, thus reducing the risk of chronic diseases linked to aging. In summary, this review provides insights into the effects and underlying mechanisms of immunosenescence, as well as its potential interventions, with particular emphasis on the relationship between aging, immunity, and nutritional factors.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Juan Chen
- Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing 100083, China; (W.Y.)
| |
Collapse
|
19
|
Zhou R, Barnes K, Gibson S, Fillmore N. Dual-edged role of SIRT1 in energy metabolism and cardiovascular disease. Am J Physiol Heart Circ Physiol 2024; 327:H1162-H1173. [PMID: 39269450 DOI: 10.1152/ajpheart.00001.2024] [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: 01/01/2024] [Revised: 08/13/2024] [Accepted: 09/06/2024] [Indexed: 09/15/2024]
Abstract
Regulation of energy metabolism is pivotal in the development of cardiovascular diseases. Dysregulation in mitochondrial fatty acid oxidation has been linked to cardiac lipid accumulation and diabetic cardiomyopathy. Sirtuin 1 (SIRT1) is a deacetylase that regulates the acetylation of various proteins involved in mitochondrial energy metabolism. SIRT1 mediates energy metabolism by directly and indirectly affecting multiple aspects of mitochondrial processes, such as mitochondrial biogenesis. SIRT1 interacts with essential mitochondrial energy regulators such as peroxisome proliferator-activated receptor-α (PPARα), PPARγ coactivator-1α, estrogen-related receptor-α, and their downstream targets. Apart from that, SIRT1 regulates additional proteins, including forkhead box protein O1 and AMP-activated protein kinase in cardiac disease. Interestingly, studies have also shown that the expression of SIRT1 plays a dual-edged role in energy metabolism. Depending on the physiological state, SIRT1 expression can be detrimental or protective. This review focuses on the molecular pathways through which SIRT1 regulates energy metabolism in cardiovascular diseases. We will review SIRT1 and discuss its role in cardiac energy metabolism and its benefits and detrimental effects in heart disease.
Collapse
Affiliation(s)
- Redemptor Zhou
- Department of Pharmaceutical Sciences, School of Pharmacy, College of Health and Human Sciences, North Dakota State University, Fargo, North Dakota, United States
| | - Kaleb Barnes
- Department of Pharmaceutical Sciences, School of Pharmacy, College of Health and Human Sciences, North Dakota State University, Fargo, North Dakota, United States
| | - Savannah Gibson
- Department of Pharmaceutical Sciences, School of Pharmacy, College of Health and Human Sciences, North Dakota State University, Fargo, North Dakota, United States
| | - Natasha Fillmore
- Department of Pharmaceutical Sciences, School of Pharmacy, College of Health and Human Sciences, North Dakota State University, Fargo, North Dakota, United States
| |
Collapse
|
20
|
Xu G, Tian C, Li Y, Fang L, Wang J, Jing Z, Li S, Chen P. Inhibition of BCAT1 expression improves recurrent miscarriage by regulating cellular dysfunction and inflammation of trophoblasts. Cell Tissue Res 2024; 398:111-121. [PMID: 39356334 DOI: 10.1007/s00441-024-03921-7] [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: 05/08/2024] [Accepted: 09/24/2024] [Indexed: 10/03/2024]
Abstract
Sustained or chronic inflammation in the placenta can result in placental insufficiency, leading to adverse reproductive outcomes such as pregnancy loss. Branched-chain amino acid transaminase 1 (BCAT1) expresses in the placenta and is involved in the pathological inflammatory response, but its role in recurrent miscarriage (RM) has not been fully investigated. In the present study, we delved into the effects of BCAT1 on trophoblast inflammation induced by lipopolysaccharide (LPS) and a mouse model of pregnancy loss induced by LPS. In vitro, after the HTR-8/SVneo cells were treated with LPS and BCATc inhibitor 2 (a selective BCAT inhibitor), the cell apoptosis was verified by TUNEL assay, and the activity of caspase-3 and caspase-9 was detected. Real-time PCR, enzyme-linked immunosorbent assay (ELISA), and immunofluorescence (IF) were used to determine the expression of inflammatory cytokines (TNF-α, IL-6, and IL-1β) and inflammasomes (NLRP3 and ASC) in LPS-treated trophoblast cells. Western blot analysis was performed to verify the expression of phospho-IκBα (p-IκBα) in cells and NF-κB p65 in the nuclei. IF staining was used to detect the nuclear translocation of NF-κB p65. The DNA binding activity of NF-κB was detected by an electrophoretic mobility shift assay (EMSA). The results demonstrated that inhibition of BCAT1 reduced trophoblast apoptosis, suppressed the release of proinflammatory cytokines, and prevented NLRP3 inflammasome activation in response to LPS. Additionally, BCAT1 inhibition blocked the activation of the NF-κB pathway in trophoblasts. This study highlights the potential therapeutic role of targeting BCAT1 in preventing adverse reproductive outcomes associated with chronic placental inflammation.
Collapse
Affiliation(s)
- Guangli Xu
- Department of Reproductive Medicine, The First Affiliated Hospital of Henan University of Chinese Medicine, 19 Renmin Road, Zhengzhou, China.
| | - Chao Tian
- Department of Reproductive Medicine, The First Affiliated Hospital of Henan University of Chinese Medicine, 19 Renmin Road, Zhengzhou, China
| | - Yanru Li
- Department of Reproductive Medicine, The First Affiliated Hospital of Henan University of Chinese Medicine, 19 Renmin Road, Zhengzhou, China
| | - Lei Fang
- Department of Reproductive Medicine, The First Affiliated Hospital of Henan University of Chinese Medicine, 19 Renmin Road, Zhengzhou, China
| | - Jing Wang
- Department of Reproductive Medicine, The First Affiliated Hospital of Henan University of Chinese Medicine, 19 Renmin Road, Zhengzhou, China
| | - Zhuqing Jing
- Department of Reproductive Medicine, The First Affiliated Hospital of Henan University of Chinese Medicine, 19 Renmin Road, Zhengzhou, China
| | - Simeng Li
- Department of Reproductive Medicine, The First Affiliated Hospital of Henan University of Chinese Medicine, 19 Renmin Road, Zhengzhou, China
| | - Ping Chen
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Henan University of Chinese Medicine, 19 Renmin Road, Zhengzhou, China.
| |
Collapse
|
21
|
Rezaeian AH, Wei W. Molecular signaling and clinical implications in the human aging-cancer cycle. Semin Cancer Biol 2024; 106-107:28-42. [PMID: 39197809 PMCID: PMC11625621 DOI: 10.1016/j.semcancer.2024.08.003] [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: 07/17/2024] [Revised: 08/07/2024] [Accepted: 08/09/2024] [Indexed: 09/01/2024]
Abstract
It is well documented that aging is associated with cancer, and likewise, cancer survivors display accelerated aging. As the number of aging individuals and cancer survivors continues to grow, it raises additional concerns across society. Therefore, unraveling the molecular mechanisms of aging in tissues is essential to developing effective therapies to fight the aging and cancer diseases in cancer survivors and cancer patients. Indeed, cellular senescence is a critical response, or a natural barrier to suppress the transition of normal cells into cancer cells, however, hypoxia which is physiologically required to maintain the stem cell niche, is increased by aging and inhibits senescence in tissues. Interestingly, oxygen restriction or hypoxia increases longevity and slows the aging process in humans, but hypoxia can also drive angiogenesis to facilitate cancer progression. In addition, cancer treatment is considered as one of the major reasons that drive cellular senescence, subsequently followed by accelerated aging. Several clinical trials have recently evaluated inhibitors to eliminate senescent cells. However, some mechanisms of aging typically can also retard cancer cell growth and progression, which might require careful strategy for better clinical outcomes. Here we describe the molecular regulation of aging and cancer in crosstalk with DNA damage and hypoxia signaling pathways in cancer patients and cancer survivors. We also update several therapeutic strategies that might be critical in reversing the cancer treatment-associated aging process.
Collapse
Affiliation(s)
- Abdol-Hossein Rezaeian
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, United States.
| | - Wenyi Wei
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, United States.
| |
Collapse
|
22
|
Elkady MA, Kabel AM, Dawood LM, Helal AI, Borg HM, Atia HA, Sabry NM, Moustafa NM, Arafa ESA, Alsufyani SE, Arab HH. Targeting the Sirtuin-1/PPAR-Gamma Axis, RAGE/HMGB1/NF-κB Signaling, and the Mitochondrial Functions by Canagliflozin Augments the Protective Effects of Levodopa/Carbidopa in Rotenone-Induced Parkinson's Disease. MEDICINA (KAUNAS, LITHUANIA) 2024; 60:1682. [PMID: 39459469 PMCID: PMC11509249 DOI: 10.3390/medicina60101682] [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: 09/03/2024] [Revised: 10/08/2024] [Accepted: 10/11/2024] [Indexed: 10/28/2024]
Abstract
Background and Objectives: Parkinson's disease (PD) is a pathological state characterized by a combined set of abnormal movements including slow motion, resting tremors, profound stiffness of skeletal muscles, or obvious abnormalities in posture and gait, together with significant behavioral changes. Until now, no single therapeutic modality was able to provide a complete cure for PD. This work was a trial to assess the immunomodulatory effects of canagliflozin with or without levodopa/carbidopa on rotenone-induced parkinsonism in Balb/c mice. Materials and Methods: In a mouse model of PD, the effect of canagliflozin with or without levodopa/carbidopa was assessed at the behavioral, biochemical, and histopathological levels. Results: The combination of levodopa/carbidopa and canagliflozin significantly mitigated the changes induced by rotenone administration regarding the behavioral tests, striatal dopamine, antioxidant status, Nrf2 content, SIRT-1/PPAR-gamma axis, RAGE/HMGB1/NF-κB signaling, and mitochondrial dysfunction; abrogated the neuroinflammatory responses, and alleviated the histomorphologic changes induced by rotenone administration relative to the groups that received either levodopa/carbidopa or canagliflozin alone. Conclusions: Canagliflozin may represent a new adjuvant therapeutic agent that may add value to the combatting effects of levodopa/carbidopa against the pathological effects of PD.
Collapse
Affiliation(s)
| | - Ahmed M. Kabel
- Pharmacology Department, Faculty of Medicine, Tanta University, Tanta 31527, Egypt;
| | - Lamees M. Dawood
- Medical Biochemistry Department, Faculty of Medicine, Tanta University, Tanta 31511, Egypt;
| | - Azza I. Helal
- Department of Histology and Cell Biology, Faculty of Medicine, Kafrelsheikh University, Kafrelsheikh 33516, Egypt;
| | - Hany M. Borg
- Physiology Department, Faculty of Medicine, Kafrelsheikh University, Kafr El-Shaikh 33516, Egypt;
| | - Hanan Abdelmawgoud Atia
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Hail, Hail 2440, Saudi Arabia;
- Department of Biochemistry and Molecular Biology, Faculty of Pharmacy, Al-Azhar University, Cairo 35527, Egypt
| | - Nesreen M. Sabry
- Clinical Oncology Department, Faculty of Medicine, Tanta University, Tanta 31527, Egypt;
| | - Nouran M. Moustafa
- Medical Microbiology & Immunology Department, Faculty of Medicine, Ain Shams University, Cairo 11566, Egypt;
- Basic Medical Science Department, College of Medicine, Dar Al Uloom University, Riyadh 13314, Saudi Arabia
| | - El-Shaimaa A. Arafa
- College of Pharmacy and Health Sciences, Ajman University, Ajman 346, United Arab Emirates;
- Center of Medical and Bio-Allied Health Sciences Research, Ajman University, Ajman 346, United Arab Emirates
| | - Shuruq E. Alsufyani
- Department of Pharmacology and Toxicology, College of Pharmacy, Taif University, P.O. Box 11099, Taif 21944, Saudi Arabia; (S.E.A.); (H.H.A.)
| | - Hany H. Arab
- Department of Pharmacology and Toxicology, College of Pharmacy, Taif University, P.O. Box 11099, Taif 21944, Saudi Arabia; (S.E.A.); (H.H.A.)
| |
Collapse
|
23
|
Li X, Li Y, Hao Q, Jin J, Wang Y. Metabolic mechanisms orchestrated by Sirtuin family to modulate inflammatory responses. Front Immunol 2024; 15:1448535. [PMID: 39372420 PMCID: PMC11449768 DOI: 10.3389/fimmu.2024.1448535] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Accepted: 09/03/2024] [Indexed: 10/08/2024] Open
Abstract
Maintaining metabolic homeostasis is crucial for cellular and organismal health throughout their lifespans. The intricate link between metabolism and inflammation through immunometabolism is pivotal in maintaining overall health and disease progression. The multifactorial nature of metabolic and inflammatory processes makes study of the relationship between them challenging. Homologs of Saccharomyces cerevisiae silent information regulator 2 protein, known as Sirtuins (SIRTs), have been demonstrated to promote longevity in various organisms. As nicotinamide adenine dinucleotide-dependent deacetylases, members of the Sirtuin family (SIRT1-7) regulate energy metabolism and inflammation. In this review, we provide an extensive analysis of SIRTs involved in regulating key metabolic pathways, including glucose, lipid, and amino acid metabolism. Furthermore, we systematically describe how the SIRTs influence inflammatory responses by modulating metabolic pathways, as well as inflammatory cells, mediators, and pathways. Current research findings on the preferential roles of different SIRTs in metabolic disorders and inflammation underscore the potential of SIRTs as viable pharmacological and therapeutic targets. Future research should focus on the development of promising compounds that target SIRTs, with the aim of enhancing their anti-inflammatory activity by influencing metabolic pathways within inflammatory cells.
Collapse
Affiliation(s)
- Xiaoqing Li
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
- Key Laboratory of Immune Response and Immunotherapy, University of Science and Technology of China, Hefei, Anhui, China
| | - Yunjia Li
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
- Key Laboratory of Immune Response and Immunotherapy, University of Science and Technology of China, Hefei, Anhui, China
| | - Quan Hao
- China Spallation Neutron Source, Dongguan, Guangdong, China
| | - Jing Jin
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
- Key Laboratory of Immune Response and Immunotherapy, University of Science and Technology of China, Hefei, Anhui, China
| | - Yi Wang
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
- Key Laboratory of Immune Response and Immunotherapy, University of Science and Technology of China, Hefei, Anhui, China
| |
Collapse
|
24
|
Metselaar DS, Meel MH, Goulding JR, du Chatinier A, Rigamonti L, Waranecki P, Geisemeyer N, de Gooijer MC, Breur M, Koster J, Veldhuijzen van Zanten SEM, Bugiani M, Franke NE, Reddy A, Wesseling P, Kaspers GJL, Hulleman E. Gemcitabine therapeutically disrupts essential SIRT1-mediated p53 repression in atypical teratoid/rhabdoid tumors. Cell Rep Med 2024; 5:101700. [PMID: 39208799 PMCID: PMC11524974 DOI: 10.1016/j.xcrm.2024.101700] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Revised: 06/13/2024] [Accepted: 08/03/2024] [Indexed: 09/04/2024]
Abstract
Atypical teratoid/rhabdoid tumors (ATRTs) are highly malignant embryonal tumors of the central nervous system with a dismal prognosis. Using a newly developed and validated patient-derived ATRT culture and xenograft model, alongside a panel of primary ATRT models, we found that ATRTs are selectively sensitive to the nucleoside analog gemcitabine. Gene expression and protein analyses indicate that gemcitabine treatment causes the degradation of sirtuin 1 (SIRT1), resulting in cell death through activation of nuclear factor κB (NF-κB) and p53. Furthermore, we discovered that gemcitabine-induced loss of SIRT1 results in a nucleus-to-cytoplasm translocation of the sonic hedgehog (SHH) signaling activator GLI2, explaining the observed additional gemcitabine sensitivity in SHH-subtype ATRT. Treatment of ATRT xenograft-bearing mice with gemcitabine resulted in a >30% increase in median survival and yielded long-term survivors in two independent patient-derived xenograft models. These findings demonstrate that ATRTs are highly sensitive to gemcitabine treatment and may form part of a future multimodal treatment strategy for ATRTs.
Collapse
Affiliation(s)
- Dennis S Metselaar
- Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands; Departments of Pediatric Oncology/Hematology, Cancer Center Amsterdam, Amsterdam University Medical Centers, Amsterdam, the Netherlands
| | - Michaël H Meel
- Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands; Departments of Pediatric Oncology/Hematology, Cancer Center Amsterdam, Amsterdam University Medical Centers, Amsterdam, the Netherlands
| | - Joshua R Goulding
- Departments of Pediatric Oncology/Hematology, Cancer Center Amsterdam, Amsterdam University Medical Centers, Amsterdam, the Netherlands
| | | | - Leyla Rigamonti
- Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands
| | - Piotr Waranecki
- Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands; Departments of Pediatric Oncology/Hematology, Cancer Center Amsterdam, Amsterdam University Medical Centers, Amsterdam, the Netherlands
| | - Neal Geisemeyer
- Hopp Children's Cancer Center (KiTZ), Heidelberg, Germany; Division of Pediatric Neuro-Oncology, German Cancer Research Center (DKFZ) and German Cancer Consortium (DKTK), Heidelberg, Germany
| | - Mark C de Gooijer
- Division of Pharmacology/Mouse Cancer Clinic, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Marjolein Breur
- Department of Pathology, Amsterdam University Medical Centers, Amsterdam, the Netherlands
| | - Jan Koster
- Department of Epidemiology and Biostatistics, Amsterdam University Medical Centers, Amsterdam, the Netherlands
| | | | - Marianna Bugiani
- Department of Pathology, Amsterdam University Medical Centers, Amsterdam, the Netherlands
| | - Niels E Franke
- Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands
| | - Alyssa Reddy
- Departments of Neurology and Pediatrics, University of California San Francisco, San Francisco, CA, USA
| | - Pieter Wesseling
- Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands; Department of Pathology, Amsterdam University Medical Centers, Amsterdam, the Netherlands
| | - Gertjan J L Kaspers
- Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands; Departments of Pediatric Oncology/Hematology, Cancer Center Amsterdam, Amsterdam University Medical Centers, Amsterdam, the Netherlands
| | - Esther Hulleman
- Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands; Departments of Pediatric Oncology/Hematology, Cancer Center Amsterdam, Amsterdam University Medical Centers, Amsterdam, the Netherlands.
| |
Collapse
|
25
|
Lu J, Zhang J, Jiang H, Hu Z, Zhang Y, He L, Yang J, Xie Y, Wu D, Li H, Zeng K, Tan P, Xiao Q, Song Z, Pan C, Bai X, Yu X. Vangl2 suppresses NF-κB signaling and ameliorates sepsis by targeting p65 for NDP52-mediated autophagic degradation. eLife 2024; 12:RP87935. [PMID: 39269442 PMCID: PMC11398866 DOI: 10.7554/elife.87935] [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] [Indexed: 09/15/2024] Open
Abstract
Van Gogh-like 2 (Vangl2), a core planar cell polarity component, plays an important role in polarized cellular and tissue morphology induction, growth development, and cancer. However, its role in regulating inflammatory responses remains elusive. Here, we report that Vangl2 is upregulated in patients with sepsis and identify Vangl2 as a negative regulator of The nuclear factor-kappaB (NF-κB) signaling by regulating the protein stability and activation of the core transcription component p65. Mice with myeloid-specific deletion of Vangl2 (Vangl2ΔM) are hypersusceptible to lipopolysaccharide (LPS)-induced septic shock. Vangl2-deficient myeloid cells exhibit enhanced phosphorylation and expression of p65, therefore, promoting the secretion of proinflammatory cytokines after LPS stimulation. Mechanistically, NF-κB signaling-induced-Vangl2 recruits E3 ubiquitin ligase PDLIM2 to catalyze K63-linked ubiquitination on p65, which serves as a recognition signal for cargo receptor NDP52-mediated selective autophagic degradation. Taken together, these findings demonstrate Vangl2 as a suppressor of NF-κB-mediated inflammation and provide insights into the crosstalk between autophagy and inflammatory diseases.
Collapse
Affiliation(s)
- Jiansen Lu
- Department of Joint Surgery, the Fifth Affiliated Hospital, Southern Medical UniversityGuangzhouChina
- Department of Immunology, School of Basic Medical Sciences, Southern Medical UniversityGuangzhouChina
| | - Jiahuan Zhang
- Department of Clinical Laboratory Medicine, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical UniversityGuangzhouChina
| | - Huaji Jiang
- Department of Immunology, School of Basic Medical Sciences, Southern Medical UniversityGuangzhouChina
- Department of Orthopaedics, Yuebei People's Hospital Affiliated to Medical College of Shantou UniversityShaoguanChina
| | - Zhiqiang Hu
- Department of Immunology, School of Basic Medical Sciences, Southern Medical UniversityGuangzhouChina
| | - Yufen Zhang
- Department of Immunology, School of Basic Medical Sciences, Southern Medical UniversityGuangzhouChina
| | - Lian He
- Department of Pharmacology, School of Medicine, Southern University of Science and TechnologyShenzhenChina
- Institute of Biosciences and Technology, College of Medicine, Texas A&M UniversityHoustonUnited States
| | - Jianwu Yang
- Department of Immunology, School of Basic Medical Sciences, Southern Medical UniversityGuangzhouChina
| | - Yingchao Xie
- Department of Immunology, School of Basic Medical Sciences, Southern Medical UniversityGuangzhouChina
| | - Dan Wu
- Department of Immunology, School of Basic Medical Sciences, Southern Medical UniversityGuangzhouChina
| | - Hongyu Li
- Department of Immunology, School of Basic Medical Sciences, Southern Medical UniversityGuangzhouChina
| | - Ke Zeng
- Department of Immunology, School of Basic Medical Sciences, Southern Medical UniversityGuangzhouChina
| | - Peng Tan
- Department of Pharmacology, School of Medicine, Southern University of Science and TechnologyShenzhenChina
- Klarman Cell Observatory, Broad Institute of MIT and HarvardCambridgeUnited States
| | - Qingyue Xiao
- Department of Cell Biology, School of Basic Medical Sciences, Southern Medical UniversityGuangzhouChina
| | - Zijing Song
- Department of Cell Biology, School of Basic Medical Sciences, Southern Medical UniversityGuangzhouChina
| | - Chenglong Pan
- Department of Joint Surgery, the Fifth Affiliated Hospital, Southern Medical UniversityGuangzhouChina
| | - Xiaochun Bai
- Department of Cell Biology, School of Basic Medical Sciences, Southern Medical UniversityGuangzhouChina
| | - Xiao Yu
- Department of Immunology, School of Basic Medical Sciences, Southern Medical UniversityGuangzhouChina
- Department of Clinical Laboratory Medicine, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical UniversityGuangzhouChina
- Guangdong Provincial Key Lab of Single Cell Technology and Application, Southern Medical UniversityGuangzhouChina
| |
Collapse
|
26
|
Chen Y, Peng S, Liang J, Wei K. SIRT1 in acute lung injury: unraveling its pleiotropic functions and therapeutic development prospects. Mol Cell Biochem 2024:10.1007/s11010-024-05111-z. [PMID: 39269678 DOI: 10.1007/s11010-024-05111-z] [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: 04/26/2024] [Accepted: 08/27/2024] [Indexed: 09/15/2024]
Abstract
Acute lung injury (ALI) is a continuum of lung changes caused by multiple lung injuries, often associated with severe complications and even death. In ALI, macrophages, alveolar epithelial cells and vascular endothelial cells in the lung are damaged to varying degrees and their function is impaired. Research in recent years has focused on the use of SIRT1 for the treatment of ALI. In this paper, we reviewed the role of SIRT1 in ALI in terms of its cellular and molecular mechanism, targeting of SIRT1 by non-coding RNAs and drug components, as well as pointing out the value of SIRT1 for clinical diagnosis and prognosis. Based on the current literature, SIRT1 exhibits diverse functionalities and possesses significant therapeutic potential. Targeting SIRT1 may provide new therapeutic ideas for the treatment of ALI.
Collapse
Affiliation(s)
- Yina Chen
- Medical School, Hunan University of Chinese Medicine, Changsha, 410208, China
| | - Shuangyan Peng
- Medical School, Hunan University of Chinese Medicine, Changsha, 410208, China
| | - Junjie Liang
- Medical School, Hunan University of Chinese Medicine, Changsha, 410208, China
| | - Ke Wei
- Medical School, Hunan University of Chinese Medicine, Changsha, 410208, China.
- Hunan Province Key Laboratory of Integrative Pathogen Biology, Hunan University of Chinese Medicine, Changsha, 410208, Hunan, China.
| |
Collapse
|
27
|
Xing H, Wu Z, Jiang K, Yuan G, Guo Z, Yu S, He S, Zhong F. FABP4 deficiency ameliorates alcoholic steatohepatitis in mice via inhibition of p53 signaling pathway. Sci Rep 2024; 14:21135. [PMID: 39256510 PMCID: PMC11387727 DOI: 10.1038/s41598-024-71311-8] [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: 04/19/2024] [Accepted: 08/27/2024] [Indexed: 09/12/2024] Open
Abstract
Fatty acid-binding protein 4 (FABP4) plays an essential role in metabolism and inflammation. However, the role of FABP4 in alcoholic steatohepatitis (ASH) remains unclear. This study aimed to investigate the function and underlying mechanisms of FABP4 in the progression of ASH. We first obtained alcoholic hepatitis (AH) datasets from the National Center for Biotechnology Information-Gene Expression Omnibus database and conducted bioinformatics analysis to identify critical genes in the FABP family. We then established ASH models of the wild-type (WT) and Fabp4-deficient (Fabp4-/-) mice to investigate the role of FABP4 in ASH. Additionally, we performed transcriptional profiling of mouse liver tissue and analyzed the results using integrative bioinformatics. The FABP4-associated signaling pathway was further verified. FABP4 was upregulated in two AH datasets and was thus identified as a critical biomarker for AH. FABP4 expression was higher in the liver tissues of patients with alcoholic liver disease and ASH mice than in the corresponding control samples. Furthermore, the Fabp4-/- ASH mice showed reduced hepatic lipid deposition and inflammation compared with the WT ASH mice. Mechanistically, Fabp4 may be involved in regulating the p53 and sirtuin-1 signaling pathways, subsequently affecting lipid metabolism and macrophage polarization in the liver of ASH mice. Our results demonstrate that Fabp4 is involved in the progression of ASH and that Fabp4 deficiency may ameliorate ASH. Therefore, FABP4 may be a potential therapeutic target for ASH treatment.
Collapse
Affiliation(s)
- Hao Xing
- Division of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, China
- Guangxi Key Laboratory of Immunology and Metabolism for Liver Diseases, Nanning, 530021, China
| | - Zhan Wu
- Division of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, China
- Guangxi Key Laboratory of Immunology and Metabolism for Liver Diseases, Nanning, 530021, China
| | - Keqing Jiang
- Division of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, China
- Guangxi Key Laboratory of Immunology and Metabolism for Liver Diseases, Nanning, 530021, China
- Key Laboratory of Early Prevention and Treatment for Regional High Frequency Tumor (Guangxi Medical University), Ministry of Education, Nanning, 530021, China
- Guangxi Key Laboratory of Basic and Clinical Application Research for Hepatobiliary Diseases, Nanning, 530021, Guangxi, China
| | - Guandou Yuan
- Division of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, China
- Guangxi Key Laboratory of Immunology and Metabolism for Liver Diseases, Nanning, 530021, China
- Key Laboratory of Early Prevention and Treatment for Regional High Frequency Tumor (Guangxi Medical University), Ministry of Education, Nanning, 530021, China
| | - Zhenya Guo
- Division of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, China
- Guangxi Key Laboratory of Immunology and Metabolism for Liver Diseases, Nanning, 530021, China
- Key Laboratory of Early Prevention and Treatment for Regional High Frequency Tumor (Guangxi Medical University), Ministry of Education, Nanning, 530021, China
| | - Shuiping Yu
- Division of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, China
- Guangxi Key Laboratory of Immunology and Metabolism for Liver Diseases, Nanning, 530021, China
- Guangxi Key Laboratory of Basic and Clinical Application Research for Hepatobiliary Diseases, Nanning, 530021, Guangxi, China
| | - Songqing He
- Division of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, China.
- Guangxi Key Laboratory of Immunology and Metabolism for Liver Diseases, Nanning, 530021, China.
- Key Laboratory of Early Prevention and Treatment for Regional High Frequency Tumor (Guangxi Medical University), Ministry of Education, Nanning, 530021, China.
- Guangxi Key Laboratory of Basic and Clinical Application Research for Hepatobiliary Diseases, Nanning, 530021, Guangxi, China.
| | - Fudi Zhong
- Division of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, China.
- Guangxi Key Laboratory of Immunology and Metabolism for Liver Diseases, Nanning, 530021, China.
- Key Laboratory of Early Prevention and Treatment for Regional High Frequency Tumor (Guangxi Medical University), Ministry of Education, Nanning, 530021, China.
- Guangxi Key Laboratory of Basic and Clinical Application Research for Hepatobiliary Diseases, Nanning, 530021, Guangxi, China.
| |
Collapse
|
28
|
Rungratanawanich W, LeFort KR, Cho YE, Li X, Song BJ. Melatonin Prevents Thioacetamide-Induced Gut Leakiness and Liver Fibrosis Through the Gut-Liver Axis via Modulating Sirt1-Related Deacetylation of Gut Junctional Complex and Hepatic Proteins. J Pineal Res 2024; 76:e13007. [PMID: 39269018 PMCID: PMC11480967 DOI: 10.1111/jpi.13007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Revised: 07/11/2024] [Accepted: 08/27/2024] [Indexed: 09/15/2024]
Abstract
Intestinal barrier dysfunction with high serum endotoxin is common in patients with liver fibrosis, but the mechanisms underlying liver fibrosis remain unclear. Melatonin is a well-recognized antioxidant and an anti-inflammatory agent that benefits multiple organs. However, the beneficial effects of melatonin on gut leakiness-associated liver fibrosis have not been systemically studied. Here, we investigated the protective mechanisms of melatonin against thioacetamide (TAA)-induced gut barrier dysfunction and hepatic fibrosis by focusing on posttranslational protein modifications through the gut-liver axis. Our results showed that gut leakiness markers, including decreased gut tight/adherens junction proteins (TJ/AJs) with increased intestinal deformation, apoptosis, and serum endotoxin, were observed early at 1 week after TAA exposure. Liver injury, apoptosis, and fibrosis were prominent at 2 and 4 weeks. Mechanistically, we found that gut TJ/AJs were hyper-acetylated, followed by ubiquitin-dependent proteolysis, leading to their degradation and gut leakiness. Gut dysbiosis, hepatic protein hyper-acetylation, and SIRT1 downregulation were also observed. Consistently, intestinal Sirt1 deficiency greatly enhanced protein hyper-acetylation, gut leakiness, endotoxemia, and liver fibrosis. Pretreatment with melatonin prevented or improved all these changes in both the gut and liver. Furthermore, melatonin blunted protein acetylation and injury in TAA-exposed T84 human intestinal and AML12 mouse liver cells. Overall, this study demonstrated novel mechanisms by which melatonin prevents gut leakiness and liver fibrosis through the gut-liver axis by attenuating the acetylation of intestinal and hepatic proteins. Thus, melatonin consumption can become a potentially safe supplement for liver fibrosis patients by preventing protein hyper-acetylation and gut leakiness.
Collapse
Affiliation(s)
- Wiramon Rungratanawanich
- Section of Molecular Pharmacology and Toxicology, National Institute on Alcohol Abuse and Alcoholism, 9000 Rockville Pike, Bethesda, MD 20892, USA
| | - Karli Rae LeFort
- Section of Molecular Pharmacology and Toxicology, National Institute on Alcohol Abuse and Alcoholism, 9000 Rockville Pike, Bethesda, MD 20892, USA
| | - Young-Eun Cho
- Department of Food and Nutrition, Andong National University, Andong, Republic of Korea
| | - Xiaoling Li
- Signal Transduction Laboratory, National Institute of Environmental Health Sciences, BG 101, Research Triangle Park, NC 27709, USA
| | - Byoung-Joon Song
- Section of Molecular Pharmacology and Toxicology, National Institute on Alcohol Abuse and Alcoholism, 9000 Rockville Pike, Bethesda, MD 20892, USA
| |
Collapse
|
29
|
Elbaz EM, Abdel Rahman AAS, El-Gazar AA, Ali BM. Protective effect of dimethyl fumarate against ethanol-provoked gastric ulcers in rats via regulation of HMGB1/TLR4/NF-κB, and PPARγ/SIRT1/Nrf2 pathways: Involvement of miR-34a-5p. Arch Biochem Biophys 2024; 759:110103. [PMID: 39053682 DOI: 10.1016/j.abb.2024.110103] [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: 04/03/2024] [Revised: 07/07/2024] [Accepted: 07/22/2024] [Indexed: 07/27/2024]
Abstract
Aberration of the gastric mucosal barrier homeostasis circuit is one of the key features linked to the onset of gastric ulcers (GU). This work aimed to inspect the gastroprotective influence of dimethyl fumarate (DMF) on ethanol-induced GU in rats and to decipher the possible mechanisms entailed. Rats were pretreated with either DMF (80 mg/kg) or omeprazole (OMP) (20 mg/kg) by oral gavage for 2 weeks. After 24 h of starvation, ethanol (5 ml/kg, oral) was employed to trigger GU in rats, while carboxymethyl cellulose (CMC) was used as a control. Ethanol notably elevated both macroscopic and microscopic gastric damage. DMF and OMP exhibited similar effects on gastric ulcer healing. DMF intervention led to a substantial improvement in gastric insults. DMF significantly reduced ethanol-triggered gastric lesions, as manifested by decreased gastric secretion, acidity, ulcer surface area percent, reduced leukocyte incursion, and increased mucus percent. DMF upregulated miR-34a-5p expression concomitant with the suppression of high mobility group box1 (HMGB1) and inflammatory responses in gastric mucosal homogenate. DMF improved GU by restoring reduced antioxidant defense mechanisms through the coactivation of nuclear factor erythroid 2-related factor-2 (Nrf2), peroxisome proliferator-activated receptor gamma (PPARγ), and sirtuin1 (SIRT1), indicating the protective role of the PPARγ/SIRT1/Nrf2 pathway. Intriguingly, DMF mitigated apoptosis in ethanol-elicited GU. Taken together, this research implies the potential for the repurposing of DMF as an innovative gastroprotective medication to reestablish the balance of the gastric mucosal barrier via the attenuation of gastric inflammation, oxidative stress, and apoptosis.
Collapse
Affiliation(s)
- Eman M Elbaz
- Department of Biochemistry, Faculty of Pharmacy, Cairo University, Cairo 11562, Egypt.
| | - Amina A S Abdel Rahman
- Department of Zoology, Faculty of Women for Arts, Science and Education, Ain Shams University, Cairo, Egypt
| | - Amira A El-Gazar
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, October 6 University, Giza 12585, Egypt
| | - Bassam Mohamed Ali
- Department of Biochemistry, Faculty of Pharmacy, October 6 University, Giza 12585, Egypt
| |
Collapse
|
30
|
Vázquez-Lizarraga R, Mendoza-Viveros L, Cid-Castro C, Ruiz-Montoya S, Carreño-Vázquez E, Orozco-Solis R. Hypothalamic circuits and aging: keeping the circadian clock updated. Neural Regen Res 2024; 19:1919-1928. [PMID: 38227516 PMCID: PMC11040316 DOI: 10.4103/1673-5374.389624] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Revised: 09/22/2023] [Accepted: 10/20/2023] [Indexed: 01/17/2024] Open
Abstract
Over the past century, age-related diseases, such as cancer, type-2 diabetes, obesity, and mental illness, have shown a significant increase, negatively impacting overall quality of life. Studies on aged animal models have unveiled a progressive discoordination at multiple regulatory levels, including transcriptional, translational, and post-translational processes, resulting from cellular stress and circadian derangements. The circadian clock emerges as a key regulator, sustaining physiological homeostasis and promoting healthy aging through timely molecular coordination of pivotal cellular processes, such as stem-cell function, cellular stress responses, and inter-tissue communication, which become disrupted during aging. Given the crucial role of hypothalamic circuits in regulating organismal physiology, metabolic control, sleep homeostasis, and circadian rhythms, and their dependence on these processes, strategies aimed at enhancing hypothalamic and circadian function, including pharmacological and non-pharmacological approaches, offer systemic benefits for healthy aging. Intranasal brain-directed drug administration represents a promising avenue for effectively targeting specific brain regions, like the hypothalamus, while reducing side effects associated with systemic drug delivery, thereby presenting new therapeutic possibilities for diverse age-related conditions.
Collapse
Affiliation(s)
| | - Lucia Mendoza-Viveros
- Instituto Nacional de Medicina Genómica (INMEGEN), México City, México
- Centro de Investigacíon sobre el Envejecimiento, Centro de Investigacíon y de Estudios Avanzados (CIE-CINVESTAV), México City, México
- Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México UNAM, México City, México
| | - Carolina Cid-Castro
- Instituto Nacional de Medicina Genómica (INMEGEN), México City, México
- Centro de Investigacíon sobre el Envejecimiento, Centro de Investigacíon y de Estudios Avanzados (CIE-CINVESTAV), México City, México
- Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México UNAM, México City, México
| | | | | | - Ricardo Orozco-Solis
- Instituto Nacional de Medicina Genómica (INMEGEN), México City, México
- Centro de Investigacíon sobre el Envejecimiento, Centro de Investigacíon y de Estudios Avanzados (CIE-CINVESTAV), México City, México
| |
Collapse
|
31
|
Campagna R, Mazzanti L, Pompei V, Alia S, Vignini A, Emanuelli M. The Multifaceted Role of Endothelial Sirt1 in Vascular Aging: An Update. Cells 2024; 13:1469. [PMID: 39273039 PMCID: PMC11394039 DOI: 10.3390/cells13171469] [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: 07/17/2024] [Revised: 08/21/2024] [Accepted: 08/28/2024] [Indexed: 09/15/2024] Open
Abstract
NAD+-dependent deacetylase sirtuin-1 (Sirt1) belongs to the sirtuins family, known to be longevity regulators, and exerts a key role in the prevention of vascular aging. By aging, the expression levels of Sirt1 decline with a severe impact on vascular function, such as the rise of endothelial dysfunction, which in turn promotes the development of cardiovascular diseases. In this context, the impact of Sirt1 activity in preventing endothelial senescence is particularly important. Given the key role of Sirt1 in counteracting endothelial senescence, great efforts have been made to deepen the knowledge about the intricate cross-talks and interactions of Sirt1 with other molecules, in order to set up possible strategies to boost Sirt1 activity to prevent or treat vascular aging. The aim of this review is to provide a proper background on the regulation and function of Sirt1 in the vascular endothelium and to discuss the recent advances regarding the therapeutic strategies of targeting Sirt1 to counteract vascular aging.
Collapse
Affiliation(s)
- Roberto Campagna
- Department of Clinical Sciences, Polytechnic University of Marche, 60100 Ancona, Italy; (V.P.); (S.A.); (A.V.); (M.E.)
| | - Laura Mazzanti
- Department of Clinical Sciences, Polytechnic University of Marche, 60100 Ancona, Italy; (V.P.); (S.A.); (A.V.); (M.E.)
- Fondazione Salesi, Ospedale G. Salesi, 60100 Ancona, Italy
| | - Veronica Pompei
- Department of Clinical Sciences, Polytechnic University of Marche, 60100 Ancona, Italy; (V.P.); (S.A.); (A.V.); (M.E.)
| | - Sonila Alia
- Department of Clinical Sciences, Polytechnic University of Marche, 60100 Ancona, Italy; (V.P.); (S.A.); (A.V.); (M.E.)
| | - Arianna Vignini
- Department of Clinical Sciences, Polytechnic University of Marche, 60100 Ancona, Italy; (V.P.); (S.A.); (A.V.); (M.E.)
- Research Center of Health Education and Health Promotion, Università Politecnica delle Marche, 60100 Ancona, Italy
| | - Monica Emanuelli
- Department of Clinical Sciences, Polytechnic University of Marche, 60100 Ancona, Italy; (V.P.); (S.A.); (A.V.); (M.E.)
| |
Collapse
|
32
|
Wang J, Hashimoto Y, Hiemori-Kondo M, Nakamoto A, Sakai T, Ye W, Abe-Kanoh N. Resveratrol and piceid enhance efferocytosis by increasing the secretion of MFG-E8 in human THP-1 macrophages. Biosci Biotechnol Biochem 2024; 88:1090-1101. [PMID: 38830798 DOI: 10.1093/bbb/zbae079] [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: 03/19/2024] [Accepted: 05/26/2024] [Indexed: 06/05/2024]
Abstract
The process of apoptotic cell clearance by phagocytes, known as efferocytosis, plays an essential role in maintaining homeostasis. Defects in efferocytosis can lead to inflammatory diseases such as atherosclerosis and autoimmune disorders. Therefore, the maintenance and promotion of efferocytosis are considered crucial for preventing these diseases. In this study, we observed that resveratrol, a representative functional food ingredient, and its glycoside, piceid, promoted efferocytosis in both human THP-1 macrophages differentiated with phorbol 12-myristate 13-acetate and peritoneal macrophages from thioglycolate-elicited mice. Resveratrol and piceid significantly increased mRNA expression and protein secretion of MFG-E8 in THP-1 macrophages. Furthermore, the activation of efferocytosis and the increment in MFG-E8 protein secretion caused by resveratrol or piceid treatment were canceled by MFG-E8 knockdown in THP-1 macrophages. In conclusion, we have demonstrated for the first time that resveratrol and piceid promote efferocytosis through the upregulation of MFG-E8 excretion in human THP-1 macrophages.
Collapse
Affiliation(s)
- Jing Wang
- Peking University Institute of Advanced Agricultural Sciences, Shandong Laboratory of Advanced Agricultural Sciences in Weifang, Weifang Key Laboratory of Grapevine Improvement and Utilization, Weifang, Shandong, China
| | - Yuki Hashimoto
- Department of Public Health and Applied Nutrition, Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima, Japan
| | - Miki Hiemori-Kondo
- Department of Nutrition, Faculty of Nutrition, University of Kochi, Kochi, Japan
| | - Akiko Nakamoto
- Department of Public Health and Applied Nutrition, Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima, Japan
| | - Tohru Sakai
- Department of Public Health and Applied Nutrition, Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima, Japan
| | - Wenxiu Ye
- Peking University Institute of Advanced Agricultural Sciences, Shandong Laboratory of Advanced Agricultural Sciences in Weifang, Weifang Key Laboratory of Grapevine Improvement and Utilization, Weifang, Shandong, China
| | - Naomi Abe-Kanoh
- Department of Food, Life and Environmental Science, Faculty of Agriculture, Yamagata University, Tsuruoka, Yamagata, Japan
| |
Collapse
|
33
|
Meng D, Yin G, Chen S, Zhang X, Yu W, Wang L, Liu H, Jiang W, Sun Y, Zhang F. Diosgenin attenuates nonalcoholic hepatic steatosis through the hepatic SIRT1/PGC-1α pathway. Eur J Pharmacol 2024; 977:176737. [PMID: 38866362 DOI: 10.1016/j.ejphar.2024.176737] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2024] [Revised: 06/04/2024] [Accepted: 06/09/2024] [Indexed: 06/14/2024]
Abstract
The prevalence of nonalcoholic fatty liver disease (NAFLD) has been increasing worldwide in recent years, causing severe economic and social burdens. Therefore, the lack of currently approved drugs for anti-NAFLD has gradually gained attention. SIRT1, as a member of the sirtuins family, is now the most widely studied in the pathophysiology of many metabolic diseases, and has great potential for preventing and treating NAFLD. Natural products such as Diosgenin (DG) have the potential to be developed as clinical drugs for the treatment of NAFLD due to their excellent multi-target therapeutic effects. In this study, we found that DG can activate the SIRT1/PGC-1α pathway and upregulate the expression of its downstream targets nuclear respiratory factor 1 (NRF1), complex IV (COX IV), mitofusin-2 (MFN2), and PPARα (perox-isome proliferator-activated receptor α) in SD rats induced by high-fat diet (HFD) and HepG2 cells caused by free fatty acids (FFAs, sodium oleate: sodium palmitate = 2:1). Conversely, the levels of dynamin-related protein 1 (DRP1) and inflammatory factors, including NF-κB p65, IL6, and TNFα, were downregulated both in vitro and in vivo. This improved mitochondrial dysfunction, fatty acid oxidation (FAO), lipid accumulation, steatosis, oxidative stress, and hepatocyte inflammation. Subsequently, we applied SIRT1 inhibitor EX527 and SIRT1 agonist SRT1720 to confirm further the necessity of activating SIRT1 for DG to exert therapeutic effects on NAFLD. In summary, these results further demonstrate the potential therapeutic role of DG as a SIRT1 natural agonist for NAFLD. (Graphical Abstracts).
Collapse
Affiliation(s)
- Decheng Meng
- The First Clinical Medical College of Shandong University of Traditional Chinese Medicine, Jinan, 250011, People's Republic of China
| | - Guoliang Yin
- The First Clinical Medical College of Shandong University of Traditional Chinese Medicine, Jinan, 250011, People's Republic of China
| | - Suwen Chen
- The First Clinical Medical College of Shandong University of Traditional Chinese Medicine, Jinan, 250011, People's Republic of China
| | - Xin Zhang
- The First Clinical Medical College of Shandong University of Traditional Chinese Medicine, Jinan, 250011, People's Republic of China
| | - Wenfei Yu
- The First Clinical Medical College of Shandong University of Traditional Chinese Medicine, Jinan, 250011, People's Republic of China
| | - Linya Wang
- Traditional Chinese Medicine College of Shandong University of Traditional Chinese Medicine, Jinan, 250011, People's Republic of China
| | - Hongshuai Liu
- Traditional Chinese Medicine College of Shandong University of Traditional Chinese Medicine, Jinan, 250011, People's Republic of China
| | - Wenying Jiang
- The First Clinical Medical College of Shandong University of Traditional Chinese Medicine, Jinan, 250011, People's Republic of China
| | - Yuqing Sun
- Traditional Chinese Medicine College of Shandong University of Traditional Chinese Medicine, Jinan, 250011, People's Republic of China
| | - Fengxia Zhang
- Department of Neurology, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, 250011, People's Republic of China.
| |
Collapse
|
34
|
Golatkar V, Bhatt LK. Artesunate attenuates isoprenaline induced cardiac hypertrophy in rats via SIRT1 inhibiting NF-κB activation. Eur J Pharmacol 2024; 977:176709. [PMID: 38843948 DOI: 10.1016/j.ejphar.2024.176709] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2024] [Revised: 06/01/2024] [Accepted: 06/03/2024] [Indexed: 06/16/2024]
Abstract
Cardiac Hypertrophy is an adaptive response of the body to physiological and pathological stimuli, which increases cardiomyocyte size, thickening of cardiac muscles and progresses to heart failure. Downregulation of SIRT1 in cardiomyocytes has been linked with the pathogenesis of cardiac hypertrophy. The present study aimed to investigate the effect of Artesunate against isoprenaline induced cardiac hypertrophy in rats via SIRT1 inhibiting NF-κB activation. Experimental cardiac hypertrophy was induced in rats by subcutaneous administration of isoprenaline (5 mg/kg) for 14 days. Artesunate was administered simultaneously for 14 days at a dose of 25 mg/kg and 50 mg/kg. Artesunate administration showed significant dose dependent attenuation in mean arterial pressure, electrocardiogram, hypertrophy index and left ventricular wall thickness compared to the disease control group. It also alleviated cardiac injury biomarkers and oxidative stress. Histological observation showed amelioration of tissue injury in the artesunate treated groups compared to the disease control group. Further, artesunate treatment increased SIRT1 expression and decreased NF-kB expression in the heart. The results of the study show the cardioprotective effect of artesunate via SIRT1 inhibiting NF-κB activation in cardiomyocytes.
Collapse
Affiliation(s)
- Vaishnavi Golatkar
- Department of Pharmacology, SVKM's Dr. Bhanuben Nanavati College of Pharmacy, Vile Parle (W), Mumbai, India
| | - Lokesh Kumar Bhatt
- Department of Pharmacology, SVKM's Dr. Bhanuben Nanavati College of Pharmacy, Vile Parle (W), Mumbai, India.
| |
Collapse
|
35
|
Keizer HG, Brands R, Oosting RS, Seinen W. A comprehensive model for the biochemistry of ageing, senescence and longevity. Biogerontology 2024; 25:615-626. [PMID: 38441836 DOI: 10.1007/s10522-024-10097-8] [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: 01/17/2024] [Accepted: 02/07/2024] [Indexed: 07/02/2024]
Abstract
Various models for ageing, each focussing on different biochemical and/or cellular pathways have been proposed. This has resulted in a complex and non-coherent portrayal of ageing. Here, we describe a concise and comprehensive model for the biochemistry of ageing consisting of three interacting signalling hubs. These are the nuclear factor kappa B complex (NFκB), controlling the innate immune system, the mammalian target for rapamycin complex, controlling cell growth, and the integrated stress responses, controlling homeostasis. This model provides a framework for most other, more detailed, biochemical pathways involved in ageing, and explains why ageing involves chronic inflammation, cellular senescence, and vulnerability to environmental stress, while starting with the spontaneous formation of advanced glycation end products. The totality of data underlying this model suggest that the gradual inhibition of the AMPK-ISR probably determines the maximal lifespan. Based on this model, anti-ageing drugs in general, are expected to show hormetic dose response curves. This complicates the process of dose-optimization. Due to its specific mechanism of action, the anti-aging drug alkaline phosphatase is an exception to this rule, because it probably exhibits saturation kinetics.
Collapse
Affiliation(s)
| | - R Brands
- AMRIF Biotechnology, Agrobusiness Park 10, 6708 PW, Wageningen, The Netherlands
| | - Ronald Sake Oosting
- AMRIF Biotechnology, Agrobusiness Park 10, 6708 PW, Wageningen, The Netherlands
| | - Willem Seinen
- AMRIF Biotechnology, Agrobusiness Park 10, 6708 PW, Wageningen, The Netherlands
- Institute for Risk Assessment Sciences (IRAS), Yalelaan 1, 3584 CL, Utrecht, The Netherlands
| |
Collapse
|
36
|
Yang L, Liu D, Jiang S, Li H, Chen L, Wu Y, Essien AE, Opoku M, Naranmandakh S, Liu S, Ru Q, Li Y. SIRT1 signaling pathways in sarcopenia: Novel mechanisms and potential therapeutic targets. Biomed Pharmacother 2024; 177:116917. [PMID: 38908209 DOI: 10.1016/j.biopha.2024.116917] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Revised: 05/29/2024] [Accepted: 06/09/2024] [Indexed: 06/24/2024] Open
Abstract
Sarcopenia is an aging-related skeletal disease characterized by decreased muscle mass, strength, and physical function, severely affecting the quality of life (QoL) of the elderly population. Sirtuin 1 (SIRT1), as a nicotinamide adenine dinucleotide (NAD+)-dependent histone deacetylases, has been reported to participate in various aging-related signaling pathways and exert protective effect on many human diseases. SIRT1 functioned as an important role in the occurrence and progression of sarcopenia through regulating key pathways related to protein homeostasis, apoptosis, mitochondrial dysfunction, insulin resistance and autophagy in skeletal muscle, including SIRT1/Forkhead Box O (FoxO), AMP-activated protein kinase (AMPK)/SIRT1/nuclear factor κB (NF-κB), SIRT1/p53, AMPK/SIRT1/peroxisome proliferator-activated receptor gamma coactivator-1α (PGC-1α), and SIRT1/live kinase B1 (LKB1)/AMPK pathways. However, the specific mechanisms of these processes have not been fully illuminated. Currently, several SIRT1-mediated interventions on sarcopenia have been preliminarily developed, such as SIRT1 activator polyphenolic compounds, exercising and calorie restriction. In this review, we summarized the predominant mechanisms of SIRT1 involved in sarcopenia and therapeutic modalities targeting the SIRT1 signaling pathways for the prevention and prognosis of sarcopenia.
Collapse
Affiliation(s)
- Luning Yang
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China; Xiangya School of Medicine, Central South University, Changsha, Hunan 410013, China
| | - Di Liu
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Shide Jiang
- Department of Orthopedics, The Central Hospital of Yongzhou, Yongzhou 425000, China
| | - Hengzhen Li
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Lin Chen
- Department of Health and Physical Education, Jianghan University, Wuhan 430056, China
| | - Yuxiang Wu
- Department of Health and Physical Education, Jianghan University, Wuhan 430056, China
| | - Anko Elijah Essien
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Michael Opoku
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Shinen Naranmandakh
- Department of chemistry, School of Arts and Sciences, National University of Mongolia, Ulaanbaatar 14201, Mongolia
| | - ShuGuang Liu
- Department of Joint Surgery, Honghui Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi, China.
| | - Qin Ru
- Department of Health and Physical Education, Jianghan University, Wuhan 430056, China.
| | - Yusheng Li
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China.
| |
Collapse
|
37
|
Lu Y, Tang X, Wang W, Yang J, Wang S. The role of deacetylase SIRT1 in allergic diseases. Front Immunol 2024; 15:1422541. [PMID: 39081309 PMCID: PMC11286408 DOI: 10.3389/fimmu.2024.1422541] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Accepted: 07/02/2024] [Indexed: 08/02/2024] Open
Abstract
The silent information regulator sirtuin 1 (SIRT1) protein is an NAD+-dependent class-III lysine deacetylase that serves as an important post-transcriptional modifier targeting lysine acetylation sites to mediate deacetylation modifications of histones and non-histone proteins. SIRT1 has been reported to be involved in several physiological or pathological processes such as aging, inflammation, immune responses, oxidative stress and allergic diseases. In this review, we summarized the regulatory roles of SIRT1 during allergic disorder progression. Furthermore, we highlight the therapeutic effects of targeting SIRT1 in allergic diseases.
Collapse
Affiliation(s)
- Yun Lu
- Department of Laboratory Medicine, Affiliated Hospital of Jiangsu University, Zhenjiang, China
- Department of Immunology, School of Medicine, Jiangsu University, Zhenjiang, China
| | - Xinyi Tang
- Department of Laboratory Medicine, Affiliated People’s Hospital, Jiangsu University, Zhenjiang, China
| | - Wenxin Wang
- Department of Laboratory Medicine, Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Jun Yang
- Department of Laboratory Medicine, Affiliated People’s Hospital, Jiangsu University, Zhenjiang, China
| | - Shengjun Wang
- Department of Laboratory Medicine, Affiliated Hospital of Jiangsu University, Zhenjiang, China
- Department of Laboratory Medicine, Affiliated People’s Hospital, Jiangsu University, Zhenjiang, China
| |
Collapse
|
38
|
Russo C, Valle MS, D’Angeli F, Surdo S, Giunta S, Barbera AC, Malaguarnera L. Beneficial Effects of Manilkara zapota-Derived Bioactive Compounds in the Epigenetic Program of Neurodevelopment. Nutrients 2024; 16:2225. [PMID: 39064669 PMCID: PMC11280255 DOI: 10.3390/nu16142225] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2024] [Revised: 07/01/2024] [Accepted: 07/09/2024] [Indexed: 07/28/2024] Open
Abstract
Gestational diet has a long-dated effect not only on the disease risk in offspring but also on the occurrence of future neurological diseases. During ontogeny, changes in the epigenetic state that shape morphological and functional differentiation of several brain areas can affect embryonic fetal development. Many epigenetic mechanisms such as DNA methylation and hydroxymethylation, histone modifications, chromatin remodeling, and non-coding RNAs control brain gene expression, both in the course of neurodevelopment and in adult brain cognitive functions. Epigenetic alterations have been linked to neuro-evolutionary disorders with intellectual disability, plasticity, and memory and synaptic learning disorders. Epigenetic processes act specifically, affecting different regions based on the accessibility of chromatin and cell-specific states, facilitating the establishment of lost balance. Recent insights have underscored the interplay between epigenetic enzymes active during embryonic development and the presence of bioactive compounds, such as vitamins and polyphenols. The fruit of Manilkara zapota contains a rich array of these bioactive compounds, which are renowned for their beneficial properties for health. In this review, we delve into the action of each bioactive micronutrient found in Manilkara zapota, elucidating their roles in those epigenetic mechanisms crucial for neuronal development and programming. Through a comprehensive understanding of these interactions, we aim to shed light on potential avenues for harnessing dietary interventions to promote optimal neurodevelopment and mitigate the risk of neurological disorders.
Collapse
Affiliation(s)
- Cristina Russo
- Section of Pathology, Department of Biomedical and Biotechnological Sciences, School of Medicine, University of Catania, 95123 Catania, Italy; (C.R.); (L.M.)
| | - Maria Stella Valle
- Section of Physiology, Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy
| | - Floriana D’Angeli
- Department of Human Sciences and Quality of Life Promotion, San Raffaele Roma Open University, 00166 Rome, Italy;
| | - Sofia Surdo
- Italian Center for the Study of Osteopathy (CSDOI), 95124 Catania, Italy;
| | - Salvatore Giunta
- Section of Anatomy, Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy;
| | - Antonio Carlo Barbera
- Section of Agronomy and Field Crops, Department of Agriculture, Food and Environment, University of Catania, 95123 Catania, Italy;
| | - Lucia Malaguarnera
- Section of Pathology, Department of Biomedical and Biotechnological Sciences, School of Medicine, University of Catania, 95123 Catania, Italy; (C.R.); (L.M.)
| |
Collapse
|
39
|
Goya L, Mateos R. Antioxidant and Anti-inflammatory Effects of Marine Phlorotannins and Bromophenols Supportive of Their Anticancer Potential. Nutr Rev 2024:nuae066. [PMID: 38894623 DOI: 10.1093/nutrit/nuae066] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/21/2024] Open
Abstract
Following the goal of optimizing nutrition, the food industry has been continuously working on food reformulation, nutritional patterns, functional foods development, and the general promotion of a healthy lifestyle. To this end, the scientific community has been increasingly investigating natural compounds that could prevent or treat chronic diseases. Phlorotannins and bromophenols are phenolic compounds particularly present in marine organisms. There is extensive evidence that shows their potential in the prevention of noncommunicable diseases, including cancer, the second cause of mortality worldwide. Numerous studies have demonstrated the anticarcinogenic activity of polyphenolic algae compounds both in cell culture and experimental animal models. Although recent reviews are also available, the present update focuses on the most recent findings related to the antioxidant/anti-inflammatory effect of seaweed phenolics, as well as their regulatory capacity for new molecular targets. Additionally, the review addresses and discusses the close link between inflammation and oxidative stress, along with their relationship with tumor onset and progression, including the most recent findings supporting this correlation. Although clinical studies are still needed to support this evidence, phlorotannins and bromophenols constitute an emerging bioactive group with high potential as chemopreventive agents and/or potential adjuvants for existing cancer therapies.
Collapse
Affiliation(s)
- Luis Goya
- Department of Metabolism and Nutrition, Institute of Food Science, Technology, and Nutrition (ICTAN-CSIC), Spanish National Research Council (CSIC), 28040 Madrid, Spain
| | - Raquel Mateos
- Department of Metabolism and Nutrition, Institute of Food Science, Technology, and Nutrition (ICTAN-CSIC), Spanish National Research Council (CSIC), 28040 Madrid, Spain
| |
Collapse
|
40
|
Shan X, Li J, Hong B, Yin H, Lu Z, Wang G, Yu N, Peng D, Wang L, Zhang C, Chen W. Comparative efficacy of sweated and non-sweated Salvia miltiorrhiza Bge. extracts on acute myocardial ischemia via regulating the PPARα/RXRα/NF-κB signaling pathway. Heliyon 2024; 10:e31923. [PMID: 38845919 PMCID: PMC11154627 DOI: 10.1016/j.heliyon.2024.e31923] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Revised: 05/23/2024] [Accepted: 05/23/2024] [Indexed: 06/09/2024] Open
Abstract
Salvia miltiorrhiza Bge. (S. miltiorrhiza) is a well-known traditional Chinese medicine for the treatment of cardiovascular diseases. The processing of S. miltiorrhiza requires the raw herbs to sweat first and then dry. The aim of this study was to investigate the anti-acute myocardial ischemia (AMI) of S. miltiorrhiza extracts (including tanshinones and phenolic acids) before and after sweating, and to further explore whether the "sweating" primary processing affected the efficacy of S. miltiorrhiza. The AMI animal model was established by subcutaneous injection of isoprenaline hydrochloride (ISO). After treatment, the cardiac function of rats was evaluated by electrocardiogram (ECG), biochemical, and histochemical analysis. Moreover, the regulation of S. miltiorrhiza extracts on the peroxisome proliferator-activated receptor α (PPARα)/retinoid X receptor α (RXRα)/nuclear transcription factor-kappa B (NF-κB) signaling pathway of rats was assessed by the Western blotting. The results showed that sweated and non-sweated S. miltiorrhiza extracts including tanshinones and phenolic acids significantly reduced ST-segment elevation in ECG and the myocardial infarction area in varying degrees. Meanwhile, sweated and non-sweated S. miltiorrhiza reversed the activities of aspartate transaminase (AST), lactic dehydrogenase (LDH), creatine kinase-MB (CK-MB), and superoxide dismutase (SOD), as well as the levels of interleukin-6 (IL-6), interleukin-10 (IL-10), and tumor necrosis factor-α (TNF-α) in AMI rats. Concurrently, the results of Western blotting revealed that S. miltiorrhiza extracts regulated the PPARα/RXRα/NF-κB signaling pathway to exert an anti-inflammatory effect. Most importantly, sweated S. miltiorrhiza tanshinones extracts are more effective than the non-sweated S. miltiorrhiza, and the anti-inflammatory efficacy of tanshinones extract was also better than that of phenolic acid extract. Although phenolic acid extracts before and after sweating were effective in anti-AMI, there was no significant difference between them. In conclusion, both tanshinones and phenolic acids extracts of sweated and non-sweated S. miltiorrhiza promote anti-oxidative stress and anti-inflammatory against AMI via regulating the PPARα/RXRα/NF-κB signaling pathway. Further, the comparations between sweated and non-sweated S. miltiorrhiza extracts indicate that sweated S. miltiorrhiza tanshinones extracts have better therapeutic effects on AMI.
Collapse
Affiliation(s)
- Xiaoxiao Shan
- School of Pharmacy, Institute of Pharmacokinetics, Anhui University of Chinese Medicine, Hefei, 230012, Anhui, China
- Center for Xin'an Medicine and Modernization of Traditional Chinese Medicine of IHM, Grand Health Research Institute of Hefei Comprehensive National Science Center, Anhui University of Chinese Medicine, Hefei, 230012, China
- Anhui Province Key Laboratory of Pharmaceutical Preparation Technology and Application, Anhui University of Chinese Medicine, Hefei, 230012, Anhui, China
- Engineering Technology Research Center of Modernized Pharmaceutics, Anhui Education Department (AUCM), Hefei, 230012, Anhui, China
- Anhui Genuine Chinese Medicinal Materials Quality Improvement Collaborative Innovation Center, Hefei, 230012, Anhui, China
| | - Junying Li
- School of Pharmacy, Institute of Pharmacokinetics, Anhui University of Chinese Medicine, Hefei, 230012, Anhui, China
- Center for Xin'an Medicine and Modernization of Traditional Chinese Medicine of IHM, Grand Health Research Institute of Hefei Comprehensive National Science Center, Anhui University of Chinese Medicine, Hefei, 230012, China
- Anhui Province Key Laboratory of Pharmaceutical Preparation Technology and Application, Anhui University of Chinese Medicine, Hefei, 230012, Anhui, China
- Engineering Technology Research Center of Modernized Pharmaceutics, Anhui Education Department (AUCM), Hefei, 230012, Anhui, China
- Anhui Genuine Chinese Medicinal Materials Quality Improvement Collaborative Innovation Center, Hefei, 230012, Anhui, China
| | - Bangzhen Hong
- School of Pharmacy, Institute of Pharmacokinetics, Anhui University of Chinese Medicine, Hefei, 230012, Anhui, China
- Center for Xin'an Medicine and Modernization of Traditional Chinese Medicine of IHM, Grand Health Research Institute of Hefei Comprehensive National Science Center, Anhui University of Chinese Medicine, Hefei, 230012, China
- Anhui Province Key Laboratory of Pharmaceutical Preparation Technology and Application, Anhui University of Chinese Medicine, Hefei, 230012, Anhui, China
- Engineering Technology Research Center of Modernized Pharmaceutics, Anhui Education Department (AUCM), Hefei, 230012, Anhui, China
- Anhui Genuine Chinese Medicinal Materials Quality Improvement Collaborative Innovation Center, Hefei, 230012, Anhui, China
| | - Huihui Yin
- School of Pharmacy, Institute of Pharmacokinetics, Anhui University of Chinese Medicine, Hefei, 230012, Anhui, China
- Center for Xin'an Medicine and Modernization of Traditional Chinese Medicine of IHM, Grand Health Research Institute of Hefei Comprehensive National Science Center, Anhui University of Chinese Medicine, Hefei, 230012, China
- Anhui Province Key Laboratory of Pharmaceutical Preparation Technology and Application, Anhui University of Chinese Medicine, Hefei, 230012, Anhui, China
- Engineering Technology Research Center of Modernized Pharmaceutics, Anhui Education Department (AUCM), Hefei, 230012, Anhui, China
- Anhui Genuine Chinese Medicinal Materials Quality Improvement Collaborative Innovation Center, Hefei, 230012, Anhui, China
| | - Ziyi Lu
- School of Pharmacy, Institute of Pharmacokinetics, Anhui University of Chinese Medicine, Hefei, 230012, Anhui, China
- Center for Xin'an Medicine and Modernization of Traditional Chinese Medicine of IHM, Grand Health Research Institute of Hefei Comprehensive National Science Center, Anhui University of Chinese Medicine, Hefei, 230012, China
- Anhui Province Key Laboratory of Pharmaceutical Preparation Technology and Application, Anhui University of Chinese Medicine, Hefei, 230012, Anhui, China
- Engineering Technology Research Center of Modernized Pharmaceutics, Anhui Education Department (AUCM), Hefei, 230012, Anhui, China
- Anhui Genuine Chinese Medicinal Materials Quality Improvement Collaborative Innovation Center, Hefei, 230012, Anhui, China
| | - Guokai Wang
- School of Pharmacy, Institute of Pharmacokinetics, Anhui University of Chinese Medicine, Hefei, 230012, Anhui, China
- Anhui Province Key Laboratory of Pharmaceutical Preparation Technology and Application, Anhui University of Chinese Medicine, Hefei, 230012, Anhui, China
- Engineering Technology Research Center of Modernized Pharmaceutics, Anhui Education Department (AUCM), Hefei, 230012, Anhui, China
| | - Nianjun Yu
- School of Pharmacy, Institute of Pharmacokinetics, Anhui University of Chinese Medicine, Hefei, 230012, Anhui, China
- Engineering Technology Research Center of Modernized Pharmaceutics, Anhui Education Department (AUCM), Hefei, 230012, Anhui, China
- Anhui Genuine Chinese Medicinal Materials Quality Improvement Collaborative Innovation Center, Hefei, 230012, Anhui, China
| | - Daiyin Peng
- School of Pharmacy, Institute of Pharmacokinetics, Anhui University of Chinese Medicine, Hefei, 230012, Anhui, China
- Engineering Technology Research Center of Modernized Pharmaceutics, Anhui Education Department (AUCM), Hefei, 230012, Anhui, China
- Anhui Genuine Chinese Medicinal Materials Quality Improvement Collaborative Innovation Center, Hefei, 230012, Anhui, China
| | - Lei Wang
- School of Pharmacy, Institute of Pharmacokinetics, Anhui University of Chinese Medicine, Hefei, 230012, Anhui, China
- Engineering Technology Research Center of Modernized Pharmaceutics, Anhui Education Department (AUCM), Hefei, 230012, Anhui, China
- Anhui Genuine Chinese Medicinal Materials Quality Improvement Collaborative Innovation Center, Hefei, 230012, Anhui, China
| | - Caiyun Zhang
- School of Pharmacy, Institute of Pharmacokinetics, Anhui University of Chinese Medicine, Hefei, 230012, Anhui, China
- Center for Xin'an Medicine and Modernization of Traditional Chinese Medicine of IHM, Grand Health Research Institute of Hefei Comprehensive National Science Center, Anhui University of Chinese Medicine, Hefei, 230012, China
- Anhui Province Key Laboratory of Pharmaceutical Preparation Technology and Application, Anhui University of Chinese Medicine, Hefei, 230012, Anhui, China
- Engineering Technology Research Center of Modernized Pharmaceutics, Anhui Education Department (AUCM), Hefei, 230012, Anhui, China
| | - Weidong Chen
- School of Pharmacy, Institute of Pharmacokinetics, Anhui University of Chinese Medicine, Hefei, 230012, Anhui, China
- Engineering Technology Research Center of Modernized Pharmaceutics, Anhui Education Department (AUCM), Hefei, 230012, Anhui, China
- Anhui Genuine Chinese Medicinal Materials Quality Improvement Collaborative Innovation Center, Hefei, 230012, Anhui, China
| |
Collapse
|
41
|
Arron HE, Marsh BD, Kell DB, Khan MA, Jaeger BR, Pretorius E. Myalgic Encephalomyelitis/Chronic Fatigue Syndrome: the biology of a neglected disease. Front Immunol 2024; 15:1386607. [PMID: 38887284 PMCID: PMC11180809 DOI: 10.3389/fimmu.2024.1386607] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Accepted: 04/11/2024] [Indexed: 06/20/2024] Open
Abstract
Myalgic Encephalomyelitis/Chronic Fatigue Syndrome (ME/CFS) is a chronic, debilitating disease characterised by a wide range of symptoms that severely impact all aspects of life. Despite its significant prevalence, ME/CFS remains one of the most understudied and misunderstood conditions in modern medicine. ME/CFS lacks standardised diagnostic criteria owing to variations in both inclusion and exclusion criteria across different diagnostic guidelines, and furthermore, there are currently no effective treatments available. Moving beyond the traditional fragmented perspectives that have limited our understanding and management of the disease, our analysis of current information on ME/CFS represents a significant paradigm shift by synthesising the disease's multifactorial origins into a cohesive model. We discuss how ME/CFS emerges from an intricate web of genetic vulnerabilities and environmental triggers, notably viral infections, leading to a complex series of pathological responses including immune dysregulation, chronic inflammation, gut dysbiosis, and metabolic disturbances. This comprehensive model not only advances our understanding of ME/CFS's pathophysiology but also opens new avenues for research and potential therapeutic strategies. By integrating these disparate elements, our work emphasises the necessity of a holistic approach to diagnosing, researching, and treating ME/CFS, urging the scientific community to reconsider the disease's complexity and the multifaceted approach required for its study and management.
Collapse
Affiliation(s)
- Hayley E. Arron
- Department of Physiological Sciences, Faculty of Science, Stellenbosch University, Stellenbosch, South Africa
| | - Benjamin D. Marsh
- MRCPCH Consultant Paediatric Neurodisability, Exeter, Devon, United Kingdom
| | - Douglas B. Kell
- Department of Physiological Sciences, Faculty of Science, Stellenbosch University, Stellenbosch, South Africa
- Department of Biochemistry and Systems Biology, Institute of Systems, Molecular and Integrative Biology, Faculty of Health and Life Sciences, University of Liverpool, Liverpool, United Kingdom
- The Novo Nordisk Foundation Centre for Biosustainability, Technical University of Denmark, Lyngby, Denmark
| | - M. Asad Khan
- Directorate of Respiratory Medicine, Manchester University Hospitals, Wythenshawe Hospital, Manchester, United Kingdom
| | - Beate R. Jaeger
- Long COVID department, Clinic St Georg, Bad Aibling, Germany
| | - Etheresia Pretorius
- Department of Physiological Sciences, Faculty of Science, Stellenbosch University, Stellenbosch, South Africa
- Department of Biochemistry and Systems Biology, Institute of Systems, Molecular and Integrative Biology, Faculty of Health and Life Sciences, University of Liverpool, Liverpool, United Kingdom
| |
Collapse
|
42
|
Kranrod J, Konkel A, Valencia R, Darwesh AM, Fischer R, Schunck WH, Seubert JM. Cardioprotective properties of OMT-28, a synthetic analog of omega-3 epoxyeicosanoids. J Biol Chem 2024; 300:107372. [PMID: 38754781 PMCID: PMC11214398 DOI: 10.1016/j.jbc.2024.107372] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Revised: 04/12/2024] [Accepted: 05/09/2024] [Indexed: 05/18/2024] Open
Abstract
OMT-28 is a metabolically robust small molecule developed to mimic the structure and function of omega-3 epoxyeicosanoids. However, it remained unknown to what extent OMT-28 also shares the cardioprotective and anti-inflammatory properties of its natural counterparts. To address this question, we analyzed the ability of OMT-28 to ameliorate hypoxia/reoxygenation (HR)-injury and lipopolysaccharide (LPS)-induced endotoxemia in cultured cardiomyocytes. Moreover, we investigated the potential of OMT-28 to limit functional damage and inflammasome activation in isolated perfused mouse hearts subjected to ischemia/reperfusion (IR) injury. In the HR model, OMT-28 (1 μM) treatment largely preserved cell viability (about 75 versus 40% with the vehicle) and mitochondrial function as indicated by the maintenance of NAD+/NADH-, ADP/ATP-, and respiratory control ratios. Moreover, OMT-28 blocked the HR-induced production of mitochondrial reactive oxygen species. Pharmacological inhibition experiments suggested that Gαi, PI3K, PPARα, and Sirt1 are essential components of the OMT-28-mediated pro-survival pathway. Counteracting inflammatory injury of cardiomyocytes, OMT-28 (1 μM) reduced LPS-induced increases in TNFα protein (by about 85% versus vehicle) and NF-κB DNA binding (by about 70% versus vehicle). In the ex vivo model, OMT-28 improved post-IR myocardial function recovery to reach about 40% of the baseline value compared to less than 20% with the vehicle. Furthermore, OMT-28 (1 μM) limited IR-induced NLRP3 inflammasome activation similarly to a direct NLRP3 inhibitor (MCC950). Overall, this study demonstrates that OMT-28 possesses potent cardio-protective and anti-inflammatory properties supporting the hypothesis that extending the bioavailability of omega-3 epoxyeicosanoids may improve their prospects as therapeutic agents.
Collapse
Affiliation(s)
- Joshua Kranrod
- Faculty of Pharmacy and Pharmaceutical Sciences, College of Health Sciences, University of Alberta, Edmonton, Alberta, Canada; Cardiovascular Research Institute, University of Alberta, Edmonton, Alberta, Canada
| | | | - Robert Valencia
- Cardiovascular Research Institute, University of Alberta, Edmonton, Alberta, Canada; Faculty of Medicine and Dentistry, Department of Pharmacology, College of Health Sciences, University of Alberta, Edmonton, Alberta, Canada
| | - Ahmed M Darwesh
- Faculty of Pharmacy and Pharmaceutical Sciences, College of Health Sciences, University of Alberta, Edmonton, Alberta, Canada
| | | | | | - John M Seubert
- Faculty of Pharmacy and Pharmaceutical Sciences, College of Health Sciences, University of Alberta, Edmonton, Alberta, Canada; Cardiovascular Research Institute, University of Alberta, Edmonton, Alberta, Canada; Faculty of Medicine and Dentistry, Department of Pharmacology, College of Health Sciences, University of Alberta, Edmonton, Alberta, Canada.
| |
Collapse
|
43
|
Chan HY, Ramasamy TS, Chung FFL, Teow SY. Role of sirtuin 1 (SIRT1) in regulation of autophagy and nuclear factor-kappa Beta (NF-ĸβ) pathways in sorafenib-resistant hepatocellular carcinoma (HCC). Cell Biochem Biophys 2024; 82:959-968. [PMID: 38466472 DOI: 10.1007/s12013-024-01247-3] [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] [Accepted: 02/29/2024] [Indexed: 03/13/2024]
Abstract
Hepatocellular carcinoma (HCC) remains a major global health problem with high incidence and mortality. Diagnosis of HCC at late stages and tumour heterogeneity in patients with different genetic profiles are known factors that complicate the disease treatment. HCC therapy becomes even more challenging in patients with drug resistance such as resistance to sorafenib, which is a common drug used in HCC patients. Sorafenib resistance can further aggravate HCC by regulating various oncogenic pathways such as autophagy and nuclear factor-kappa Beta (NF-ĸβ) signalling. Sirtuin 1 (SIRT1), is a nicotinamide adenosine dinucleotide (NAD)-dependent histone deacetylases that regulates various metabolic and oncogenic events such as cell survival, apoptosis, autophagy, tumourigenesis, metastasis and drug resistance in various cancers, but its role in HCC, particularly in sorafenib resistance is underexplored. In this study, we generated sorafenib-resistant HepG2 and Huh-7 liver cancer cell models to investigate the role of SIRT1 and its effect on autophagy and nuclear factor-kappa Beta (NF-ĸβ) signalling pathways. Western blot analysis showed increased SIRT1, altered autophagy pathway and activated NF-ĸβ signalling in sorafenib-resistant cells. SIRT1-silenced HCC cells demonstrated down-regulated autophagy in both parental and chemoresistant cells. This may occur through the deacetylation of key autophagy molecules such as FOXO3, beclin 1, ATGs and LC3 by SIRT1, highlighting the role of SIRT1 in autophagy induction. Silencing of SIRT1 also resulted in activated NF-ĸβ signalling. This is because SIRT1 failed to deacetylate p65 subunit of NF-κB, translocate the NF-κB from nucleus to cytoplasm, and suppress NF-κB activity due to the silencing. Hence, the NF-κB transcriptional activity was restored. These findings summarize the role of SIRT1 in autophagy/NF-ĸβ regulatory axis, with a similar trend observed in both parental and sorafenib-resistant cells. The present work promotes a better understanding of the role of SIRT1 in autophagy and NF-ĸβ signalling in HCC and sorafenib-resistant HCC. As some key proteins in these pathways are potential therapeutic targets, a better understanding of SIRT1/autophagy/NF-ĸβ axis could further improve the therapeutic strategies against HCC.
Collapse
Affiliation(s)
- Hui-Yin Chan
- Department of Medical Sciences, School of Medical and Life Sciences, Sunway University, Jalan Universiti, 47500 Subang Jaya, Bandar, Sunway, Selangor Darul Ehsan, Malaysia
| | - Thamil Selvee Ramasamy
- Stem Cell Biology Laboratory, Department of Molecular Medicine, Faculty of Medicine, Universiti Malaya, 50603, Kuala Lumpur, Malaysia
| | - Felicia Fei-Lei Chung
- Department of Medical Sciences, School of Medical and Life Sciences, Sunway University, Jalan Universiti, 47500 Subang Jaya, Bandar, Sunway, Selangor Darul Ehsan, Malaysia
| | - Sin-Yeang Teow
- Department of Biology, College of Science, Mathematics and Technology, Wenzhou-Kean University, 88 Daxue Road, Ouhai, Wenzhou, 325060, Zhejiang Provinve, China.
- Wenzhou Municipal Key Lab for Applied Biomedical and Biopharmaceutical Informatics, Ouhai, Wenzhou, 325060, Zhejiang Province, China.
- Zhejiang Bioinformatics International Science and Technology Cooperation Center, Ouhai, Wenzhou, 325060, Zhejiang Province, China.
- Dorothy and George Hennings College of Science, Mathematics and Technology, Kean University, 1000 Morries Ave, Union, NJ, 07083, USA.
| |
Collapse
|
44
|
Xu M, Qian LH, Wang JX, He ZY, Ling XY, Wang WH, Wang JW, Hu Y, Gong MJ. Rutaecarpine Alleviates Early Brain Injury-Induced Inflammatory Response Following Subarachnoid Hemorrhage via SIRT6/NF-[Formula: see text]B Pathway. THE AMERICAN JOURNAL OF CHINESE MEDICINE 2024; 52:799-819. [PMID: 38752843 DOI: 10.1142/s0192415x24500320] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/31/2024]
Abstract
Subarachnoid hemorrhage (SAH), a specific subtype of cerebrovascular accident, is characterized by the extravasation of blood into the interstice between the brain and its enveloping delicate tissues. This pathophysiological phenomenon can precipitate an early brain injury (EBI), which is characterized by inflammation and neuronal death. Rutaecarpine (Rut), a flavonoid compound discovered in various plants, has been shown to have protective effects against SAH-induced cerebral insult in rodent models. In our study, we used a rodent SAH model to evaluate the effect of Rut on EBI and investigated the effect of Rut on the inflammatory response and its regulation of SIRT6 expression in vitro. We found that Rut exerts a protective effect on EBI in SAH rats, which is partly due to its ability to inhibit the inflammatory response. Notably, Rut up-regulated Sirtuin 6 (SIRT6) expression, leading to an increase in H3K9 deacetylation and inhibition of nuclear factor-kappa B (NF-[Formula: see text]B) transcriptional activation, thereby mediating the inflammatory response. In addition, further data showed that SIRT6 was proven to mediate the regulation of Rut on the microglial inflammatory response. These findings highlight the importance of SIRT6 in the regulation of inflammation and suggest a potential mechanism for the protective effect of Rut on EBI. In summary, Rut may have the potential to prevent and treat SAH-induced brain injury by interacting with SIRT6. Our findings may provide a new therapeutic strategy for the treatment of SAH-induced EBI.
Collapse
Affiliation(s)
- Min Xu
- Department of Neurosurgery, Kunshan Hospital of Traditional Chinese Medicine, Kunshan Affiliated Hospital of Nanjing University of Chinese Medicine, Kunshan 215300, Jiangsu Province, P. R. China
| | - Li-Hui Qian
- School of Medicine, Nanjing University of Chinese Medicine 210023, Nanjing, P. R. China
| | - Jun-Xiang Wang
- Department of Neurosurgery, Changshu No. 2 People's Hospital, Affiliated Changshu Hospital of Nantong University 215500, Jiangsu Province, P. R. China
| | - Zi-Yang He
- Department of Neurosurgery, Kunshan Hospital of Traditional Chinese Medicine, Kunshan Affiliated Hospital of Nanjing University of Chinese Medicine, Kunshan 215300, Jiangsu Province, P. R. China
| | - Xiao-Yang Ling
- Department of Neurosurgery, Kunshan Hospital of Traditional Chinese Medicine, Kunshan Affiliated Hospital of Nanjing University of Chinese Medicine, Kunshan 215300, Jiangsu Province, P. R. China
| | - Wen-Hua Wang
- Department of Neurosurgery, Kunshan Hospital of Traditional Chinese Medicine, Kunshan Affiliated Hospital of Nanjing University of Chinese Medicine, Kunshan 215300, Jiangsu Province, P. R. China
| | - Jin-Wen Wang
- School of Integrated Chinese and Western Medicine, Nanjing University of Chinese Medicine 210023, Nanjing, P. R. China
| | - Yue Hu
- School of Integrated Chinese and Western Medicine, Nanjing University of Chinese Medicine 210023, Nanjing, P. R. China
- Shen Chun-Ti Nation-Famous Experts Studio for Traditional Chinese Medicine Inheritance, Changzhou TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Changzhou 213003, Jiangsu, P. R. China
- Department of Neurology, Nanjing Hospital of Chinese Medicine affiliated to Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210001, P. R. China
| | - Ming-Jie Gong
- Department of Neurosurgery, Changshu No. 2 People's Hospital, Affiliated Changshu Hospital of Nantong University 215500, Jiangsu Province, P. R. China
| |
Collapse
|
45
|
Liu Y, Liu H, Shao Q, Shi H, Cheng F, Wang X. Majie Cataplasm Alleviates Asthma by Regulating Th1/Th2/Treg/Th17 Balance. Int Arch Allergy Immunol 2024; 185:900-909. [PMID: 38749400 DOI: 10.1159/000538597] [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: 01/12/2024] [Accepted: 03/26/2024] [Indexed: 09/04/2024] Open
Abstract
INTRODUCTION T cells play a critical role in inflammatory diseases. The aim of the present study was to investigate the effects of Majie cataplasm (MJC) on asthma and to propose a possible mechanism involved in this process. METHODS Airway inflammation, infiltration of inflammatory cells, levels of interleukin (IL)-4, IL-10, IL-17, and interferon (IFN)-γ, levels of Th2, Treg, Th17, and Th1 cells, and the expressions of IL-4, IL-10, IL-17, IFN-γ, GATA binding protein 3 (GATA-3), Foxp3, RAR-related orphan receptor gamma (RORγt), and T-bet were detected. RESULT MJC treatment reduced lung airway resistance and inflammatory infiltration in lung tissues. MJC treatment also reduced the numbers of eosinophils and neutrophils in the blood and bronchoalveolar lavage fluid (BALF). The levels of IL-4 and IL-17 in the blood, BALF, and lungs were suppressed by MJC, and IFN-γ and IL-10 were increased. Furthermore, MJC suppressed the percentage of Th2 and Th17 and increased the percentage of Th1 and Treg in spleen cells. In addition, MJC can inhibit asthma by increasing expressions of IFN-γ, IL-10, T-bet, and Foxp3, as well as decreasing expressions of IL-4, IL-17, GATA-3, and RORγt. CONCLUSION MJC may improve airway hyperresponsiveness and inflammation by regulating Th1/Th2/Treg/Th17 balance in ovalbumin-induced rats. And MJC may be a new source of anti-asthma drugs.
Collapse
Affiliation(s)
- Yuanjun Liu
- Beijing Key Laboratory, Traditional Chinese Medicine School, Beijing University of Chinese Medicine, Beijing, China
| | - Haixia Liu
- Disease-Syndrome Research Center, China Academy of Chinese Medical Sciences Institute of Basic Theory for Chinese Medicine, Beijing, China,
| | - Qi Shao
- Beijing Key Laboratory, Traditional Chinese Medicine School, Beijing University of Chinese Medicine, Beijing, China
| | - Hanfen Shi
- Beijing Key Laboratory, Traditional Chinese Medicine School, Beijing University of Chinese Medicine, Beijing, China
| | - Fafeng Cheng
- Beijing Key Laboratory, Traditional Chinese Medicine School, Beijing University of Chinese Medicine, Beijing, China
| | - Xueqian Wang
- Beijing Key Laboratory, Traditional Chinese Medicine School, Beijing University of Chinese Medicine, Beijing, China
| |
Collapse
|
46
|
Perico L, Remuzzi G, Benigni A. Sirtuins in kidney health and disease. Nat Rev Nephrol 2024; 20:313-329. [PMID: 38321168 DOI: 10.1038/s41581-024-00806-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/04/2024] [Indexed: 02/08/2024]
Abstract
Sirtuins (SIRTs) are putative regulators of lifespan in model organisms. Since the initial discovery that SIRTs could promote longevity in nematodes and flies, the identification of additional properties of these proteins has led to understanding of their roles as exquisite sensors that link metabolic activity to oxidative states. SIRTs have major roles in biological processes that are important in kidney development and physiological functions, including mitochondrial metabolism, oxidative stress, autophagy, DNA repair and inflammation. Furthermore, altered SIRT activity has been implicated in the pathophysiology and progression of acute and chronic kidney diseases, including acute kidney injury, diabetic kidney disease, chronic kidney disease, polycystic kidney disease, autoimmune diseases and renal ageing. The renoprotective roles of SIRTs in these diseases make them attractive therapeutic targets. A number of SIRT-activating compounds have shown beneficial effects in kidney disease models; however, further research is needed to identify novel SIRT-targeting strategies with the potential to treat and/or prevent the progression of kidney diseases and increase the average human healthspan.
Collapse
Affiliation(s)
- Luca Perico
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Bergamo, Italy
| | - Giuseppe Remuzzi
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Bergamo, Italy
| | - Ariela Benigni
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Bergamo, Italy.
| |
Collapse
|
47
|
Chen T, Huang X, Zhao YX, Zhou ZW, Zhou WS. NEAT1 inhibits the angiogenic activity of cerebral arterial endothelial cells by inducing the M1 polarization of microglia through the AMPK signaling pathway. Cell Mol Biol Lett 2024; 29:62. [PMID: 38684954 PMCID: PMC11059773 DOI: 10.1186/s11658-024-00579-5] [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: 12/20/2023] [Accepted: 04/15/2024] [Indexed: 05/02/2024] Open
Abstract
BACKGROUND Enhancing angiogenesis may be an effective strategy to promote functional recovery after ischemic stroke. Inflammation regulates angiogenesis. Microglia are crucial cells that initiate inflammatory responses after various brain injuries. Long noncoding RNA nuclear paraspeckle assembly transcript 1 (NEAT1) plays a role in regulating brain injury. This study aimed to explore the effects of NEAT1-regulated microglial polarization on the neovascularization capacity of cerebrovascular endothelial cells and the underlying molecular regulatory mechanisms. METHODS Mouse cerebral arterial endothelial cells (mCAECs) were co-cultured with BV-2 cells in different groups using a Transwell system. NEAT1 expression levels were measured by fluorescence quantitative reverse transcription PCR. Levels of IL-1β, IL-6, TNF-α, Arg-1, IL-4, and IL-10 were determined using ELISA. Expression levels of CD86 and CD163 were detected by immunofluorescence. The neovascularization capacity of mCAECs was assessed using CCK-8, Transwell, Transwell-matrigel, and tube formation assays. Label-free quantification proteomics was carried out to identify differentially expressed proteins. Protein levels were measured by Western blotting. RESULTS NEAT1 overexpression induced M1 polarization in BV-2 cells, whereas NEAT1 knockdown blocked lipopolysaccharide-induced M1 polarization in microglia. NEAT1-overexpressing BV-2 cells suppressed the angiogenic ability of mCAECs, and NEAT1-knocking BV-2 cells promoted the angiogenic ability of mCAECs under lipopolysaccharide treatment. Label-free quantitative proteomic analysis identified 144 upregulated and 131 downregulated proteins that were induced by NEAT1 overexpression. The AMP-activated protein kinase (AMPK) signaling pathway was enriched in the Kyoto Encyclopedia of Genes and Genomes analysis of the differentially expressed proteins. Further verification showed that NEAT1 inactivated the AMPK signaling pathway. Moreover, the AMPK activator 5-aminoimidazole-4-carboxamide ribonucleotide reversed the effect of NEAT1 on BV-2 polarization and the regulatory effect of NEAT1-overexpressing BV-2 cells on the angiogenic ability of mCAECs. CONCLUSIONS NEAT1 inhibits the angiogenic activity of mCAECs by inducing M1 polarization of BV-2 cells through the AMPK signaling pathway. This study further clarified the impact and mechanism of NEAT1 on microglia and the angiogenic ability of cerebrovascular endothelial cells.
Collapse
Affiliation(s)
- Ting Chen
- Department of Neurology, Hunan Provincial People's Hospital (The First Affiliated Hospital of Hunan Normal University), Changsha, 410005, China
| | - Xin Huang
- Department of Neurology, Hunan Provincial People's Hospital (The First Affiliated Hospital of Hunan Normal University), Changsha, 410005, China
| | - Yi-Xuan Zhao
- Department of Neurology, Hunan Provincial People's Hospital (The First Affiliated Hospital of Hunan Normal University), Changsha, 410005, China
| | - Zhi-Wen Zhou
- Department of Neurology, Hunan Provincial People's Hospital (The First Affiliated Hospital of Hunan Normal University), Changsha, 410005, China.
| | - Wen-Sheng Zhou
- Department of Neurology, Hunan Provincial People's Hospital (The First Affiliated Hospital of Hunan Normal University), Changsha, 410005, China.
| |
Collapse
|
48
|
Tong J, Song J, Zhang W, Zhai J, Guan Q, Wang H, Liu G, Zheng C. When DNA-damage responses meet innate and adaptive immunity. Cell Mol Life Sci 2024; 81:185. [PMID: 38630271 PMCID: PMC11023972 DOI: 10.1007/s00018-024-05214-2] [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: 11/04/2023] [Revised: 03/17/2024] [Accepted: 03/18/2024] [Indexed: 04/19/2024]
Abstract
When cells proliferate, stress on DNA replication or exposure to endogenous or external insults frequently results in DNA damage. DNA-Damage Response (DDR) networks are complex signaling pathways used by multicellular organisms to prevent DNA damage. Depending on the type of broken DNA, the various pathways, Base-Excision Repair (BER), Nucleotide Excision Repair (NER), Mismatch Repair (MMR), Homologous Recombination (HR), Non-Homologous End-Joining (NHEJ), Interstrand Crosslink (ICL) repair, and other direct repair pathways, can be activated separately or in combination to repair DNA damage. To preserve homeostasis, innate and adaptive immune responses are effective defenses against endogenous mutation or invasion by external pathogens. It is interesting to note that new research keeps showing how closely DDR components and the immune system are related. DDR and immunological response are linked by immune effectors such as the cyclic GMP-AMP synthase (cGAS)-Stimulator of Interferon Genes (STING) pathway. These effectors act as sensors of DNA damage-caused immune response. Furthermore, DDR components themselves function in immune responses to trigger the generation of inflammatory cytokines in a cascade or even trigger programmed cell death. Defective DDR components are known to disrupt genomic stability and compromise immunological responses, aggravating immune imbalance and leading to serious diseases such as cancer and autoimmune disorders. This study examines the most recent developments in the interaction between DDR elements and immunological responses. The DDR network's immune modulators' dual roles may offer new perspectives on treating infectious disorders linked to DNA damage, including cancer, and on the development of target immunotherapy.
Collapse
Affiliation(s)
- Jie Tong
- College of Life Science, Hebei University, Baoding, 071002, China
- Institute of Life Science and Green Development, Hebei University, Baoding, 071002, China
| | - Jiangwei Song
- Beijing Key Laboratory for Prevention and Control of Infectious Diseases in Livestock and Poultry, Institute of Animal Husbandry and Veterinary Medicine, Beijing Academy of Agriculture and Forestry Sciences, Beijing, 100089, China
| | - Wuchao Zhang
- College of Veterinary Medicine, Hebei Agricultural University, Baoding, 071000, China
| | - Jingbo Zhai
- Key Laboratory of Zoonose Prevention and Control at Universities of Inner Mongolia Autonomous Region, Medical College, Inner Mongolia Minzu University, Tongliao, 028000, China
| | - Qingli Guan
- The Affiliated Hospital of Chinese PLA 80th Group Army, Weifang, 261000, China
| | - Huiqing Wang
- Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, 610041, China.
| | - Gentao Liu
- Department of Oncology, Tenth People's Hospital Affiliated to Tongji University & Cancer Center, Tongji University School of Medicine, Shanghai, 20000, China.
| | - Chunfu Zheng
- Department of Microbiology, Immunology and Infectious Diseases, University of Calgary, Calgary, AB, Canada.
| |
Collapse
|
49
|
Zhu Q, Wan L, Huang H, Liao Z. IL-1β, the first piece to the puzzle of sepsis-related cognitive impairment? Front Neurosci 2024; 18:1370406. [PMID: 38665289 PMCID: PMC11043581 DOI: 10.3389/fnins.2024.1370406] [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: 01/14/2024] [Accepted: 04/02/2024] [Indexed: 04/28/2024] Open
Abstract
Sepsis is a leading cause of death resulting from an uncontrolled inflammatory response to an infectious agent. Multiple organ injuries, including brain injuries, are common in sepsis. The underlying mechanism of sepsis-associated encephalopathy (SAE), which is associated with neuroinflammation, is not yet fully understood. Recent studies suggest that the release of interleukin-1β (IL-1β) following activation of microglial cells plays a crucial role in the development of long-lasting neuroinflammation after the initial sepsis episode. This review provides a comprehensive analysis of the recent literature on the molecular signaling pathways involved in microglial cell activation and interleukin-1β release. It also explores the physiological and pathophysiological role of IL-1β in cognitive function, with a particular focus on its contribution to long-lasting neuroinflammation after sepsis. The findings from this review may assist healthcare providers in developing novel interventions against SAE.
Collapse
Affiliation(s)
- Qing Zhu
- Department of Anesthesiology, Key Laboratory of Birth Defects and Related Diseases of Women and Children, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Li Wan
- Department of Medical Genetics/Prenatal Diagnostic Center Nursing and Key Laboratory of Birth Defects and Related Diseases of Women and Children, West China Second University Hospital of Sichuan University, Chengdu, China
| | - Han Huang
- Department of Anesthesiology, Key Laboratory of Birth Defects and Related Diseases of Women and Children, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Zhimin Liao
- Department of Anesthesiology, Key Laboratory of Birth Defects and Related Diseases of Women and Children, West China Second University Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
50
|
Yang K, Zeng L, Li Y, Wu L, Xiang W, Wu X, Wang G, Bao T, Huang S, Yu R, Zhang G, Liu H. Uncovering the pharmacological mechanism of Shou Tai Wan on recurrent spontaneous abortion: A integrated pharmacology strategy-based research. JOURNAL OF ETHNOPHARMACOLOGY 2024; 323:117589. [PMID: 38104875 DOI: 10.1016/j.jep.2023.117589] [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: 05/17/2023] [Revised: 12/04/2023] [Accepted: 12/11/2023] [Indexed: 12/19/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Shou Tai Wan (STW), a traditional Chinese medicine formula, has been historically used for the treatment of recurrent spontaneous abortion (RSA). Despite its long-standing usage, the exact mechanism underlying the therapeutic effects of STW remains unclear in the existing literature. AIMS OF THIS STUDY To explore the Pharmacological Mechanism of STW on RSA. METHODS A network pharmacological methodology was utilized to predict the active compounds and potential targets of STW, collect the RSA targets and other human proteins of STW, and analyze the STW related networks. The animal experiments were also performed to validate the effect of STW on RSA. RESULTS The results of network analysis showed that STW may regulate PI3K/AKT, MAPK, FoxO signaling pathways and so on. Animal experiment established the RSA model with CBA/J × DBA/2 mice. It was found that STW can reduce the embryo absorption rate of RSA group (p < 0.05) and balance the expression of Th 1/Th2 type cytokines compared with the model group. After 14 days of administration, the decidual and placental tissues were taken and the CD4+ T cells were isolated, and the phosphorylation level of signaling pathway was detected by Springbio720 antibody microarray. This experiment found that STW can significantly up-regulate the phosphorylation levels of STAT3 and STAT6 proteins in the STAT signaling pathway, and down-regulating the phosphorylation level of STAT1 protein. STW also significantly up-regulated the phosphorylation levels of Raf1, A-Raf, Ask1, Mek1, Mek2, JKK1, ERK1, ERK2, c-fos, c-Jun and CREB proteins in the MAPK signaling pathway, and down-regulate the phosphorylation levels of MEK6 and IKKb proteins. Compared with the RSA group, the STW group increased the expression levels of ERK1/2 mRNA and proteins and p-ERK1/2 proteins, and there was a statistical difference (p < 0.05). This is consistent with the chip results. CONCLUSION STW may achieve therapeutic effects by interfering with the signaling pathways, biological processes and targets discovered in this study. It provides a new perspective for revealing the immunological mechanism of STW in the treatment of RSA, and also provides a theoretical basis for the clinical use of STW in the treatment of RSA.
Collapse
Affiliation(s)
- Kailin Yang
- Hunan University of Chinese Medicine, Changsha, Hunan Province, China
| | - Liuting Zeng
- Hunan University of Chinese Medicine, Changsha, Hunan Province, China
| | - Yuwei Li
- Hunan University of Science and Technology, Xiangtan, China
| | - Lingyu Wu
- Hunan University of Chinese Medicine, Changsha, Hunan Province, China
| | - Wang Xiang
- The First People's Hospital Changde City, Changde City, China
| | - Xiaolan Wu
- Hunan University of Chinese Medicine, Changsha, Hunan Province, China
| | - Guiyun Wang
- Hunan University of Chinese Medicine, Changsha, Hunan Province, China
| | - Tingting Bao
- Institute of Metabolic Diseases, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, No.5 BeiXianGe Street, Xicheng District, Beijing 100053, China
| | - Shanshan Huang
- Hunan University of Chinese Medicine, Changsha, Hunan Province, China
| | - Rong Yu
- Hunan University of Chinese Medicine, Changsha, Hunan Province, China
| | - Guomin Zhang
- Hunan University of Chinese Medicine, Changsha, Hunan Province, China.
| | - Huiping Liu
- Hunan University of Chinese Medicine, Changsha, Hunan Province, China.
| |
Collapse
|