1
|
Tarantino G, Citro V. Crosstalk Between the Spleen and Other Organs/Systems: Downstream Signaling Events. IMMUNO 2024; 4:479-501. [DOI: 10.3390/immuno4040030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2025] Open
Abstract
The aim of this review was to gather pieces of information from available critically evaluated published articles concerning any interplay in which the spleen could be involved. For many years, the spleen has been alleged as an unnecessary biological structure, even though splenomegaly is an objective finding of many illnesses. Indeed, the previous opinion has been completely changed. In fact, the spleen is not a passive participant in or a simple bystander to a relationship that exists between the immune system and other organs. Recently, it has been evidenced in many preclinical and clinical studies that there are close associations between the spleen and other parts of the body, leading to various spleen–organ axes. Among them, the gut–spleen axis, the liver–spleen axis, the gut–spleen–skin axis, the brain–spleen axis, and the cardio-splenic axis are the most explored and present in the medical literature. Such recent sources of evidence have led to revolutionary new ideas being developed about the spleen. What is more, these observations may enable the identification of novel therapeutic strategies targeted at various current diseases. The time has come to make clear that the spleen is not a superfluous body part, while health system operators and physicians should pay more attention to this organ. Indeed, much work remains to be performed to assess further roles that this biological structure could play.
Collapse
Affiliation(s)
| | - Vincenzo Citro
- Department of General Medicine, Umberto I Hospital, 84014 Nocera Inferiore, Italy
| |
Collapse
|
2
|
Bonomi RE, Riordan W, Gelovani JG. The Structures, Functions, and Roles of Class III HDACs (Sirtuins) in Neuropsychiatric Diseases. Cells 2024; 13:1644. [PMID: 39404407 PMCID: PMC11476333 DOI: 10.3390/cells13191644] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Revised: 09/29/2024] [Accepted: 09/30/2024] [Indexed: 10/19/2024] Open
Abstract
Over the past two decades, epigenetic regulation has become a rapidly growing and influential field in biology and medicine. One key mechanism involves the acetylation and deacetylation of lysine residues on histone core proteins and other critical proteins that regulate gene expression and cellular signaling. Although histone deacetylases (HDACs) have received significant attention, the roles of individual HDAC isoforms in the pathogenesis of psychiatric diseases still require further research. This is particularly true with regard to the sirtuins, class III HDACs. Sirtuins have unique functional activity and significant roles in normal neurophysiology, as well as in the mechanisms of addiction, mood disorders, and other neuropsychiatric abnormalities. This review aims to elucidate the differences in catalytic structure and function of the seven sirtuins as they relate to psychiatry.
Collapse
Affiliation(s)
- Robin E. Bonomi
- Department of Psychiatry, Yale University, New Haven, CT 06511, USA;
| | - William Riordan
- Department of Psychiatry, Yale University, New Haven, CT 06511, USA;
| | - Juri G. Gelovani
- College of Medicine and Health Sciences, Office of the Provost, United Arab Emirates University, Al Ain P.O. Box 15551, United Arab Emirates;
- Department of Biomedical Engineering, College of Engineering and School of Medicine, Wayne State University, Detroit, MI 48201, USA
- Department of Radiology, Division of Nuclear Medicine, Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand
| |
Collapse
|
3
|
Tarantino G, Citro V. What are the common downstream molecular events between alcoholic and nonalcoholic fatty liver? Lipids Health Dis 2024; 23:41. [PMID: 38331795 PMCID: PMC10851522 DOI: 10.1186/s12944-024-02031-1] [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: 11/05/2023] [Accepted: 01/25/2024] [Indexed: 02/10/2024] Open
Abstract
Liver fat storage, also called hepatic steatosis, is increasingly common and represents a very frequent diagnosis in the medical field. Excess fat is not without consequences. In fact, hepatic steatosis contributes to the progression toward liver fibrosis. There are two main types of fatty liver disease, alcoholic fatty liver disease (AFLD) and nonalcoholic fatty liver disease (NAFLD). Although AFLD and NAFLD are similar in their initial morphological features, both conditions involve the same evolutive forms. Moreover, there are various common mechanisms underlying both diseases, including alcoholic liver disease and NAFLD, which are commonalities. In this Review, the authors explore similar downstream signaling events involved in the onset and progression of the two entities but not completely different entities, predominantly focusing on the gut microbiome. Downstream molecular events, such as the roles of sirtuins, cytokeratins, adipokines and others, should be considered. Finally, to complete the feature, some new tendencies in the therapeutic approach are presented.
Collapse
Affiliation(s)
| | - Vincenzo Citro
- Department of General Medicine, Umberto I Hospital, Nocera Inferiore, SA, 84014, Italy
| |
Collapse
|
4
|
Shokeen K, Kumar S. Newcastle disease virus regulates its replication by instigating oxidative stress-driven Sirtuin 7 production. J Gen Virol 2024; 105. [PMID: 38376490 DOI: 10.1099/jgv.0.001961] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/21/2024] Open
Abstract
Reactive oxygen species (ROS) accumulation inside the cells instigates oxidative stress, activating stress-responsive genes. The viral strategies for promoting stressful conditions and utilizing the induced host proteins to enhance their replication remain elusive. The present work investigates the impact of oxidative stress responses on Newcastle disease virus (NDV) pathogenesis. Here, we show that the progression of NDV infection varies with intracellular ROS levels. Additionally, the results demonstrate that NDV infection modulates the expression of oxidative stress-responsive genes, majorly sirtuin 7 (SIRT7), a NAD+-dependent deacetylase. The modulation of SIRT7 protein, both through overexpression and knockdown, significantly impacts the replication dynamics of NDV in DF-1 cells. The activation of SIRT7 is found to be associated with the positive regulation of cellular protein deacetylation. Lastly, the results suggested that NDV-driven SIRT7 alters NAD+ metabolism in vitro and in ovo. We concluded that the elevated expression of NDV-mediated SIRT7 protein with enhanced activity metabolizes the NAD+ to deacetylase the host proteins, thus contributing to high virus replication.
Collapse
Affiliation(s)
- Kamal Shokeen
- Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati, Assam 781039, India
| | - Sachin Kumar
- Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati, Assam 781039, India
| |
Collapse
|
5
|
Hao Y, Li W. Regulatory factor X7 Represses Ox-LDL-Induced Proliferation and Migration of VSMCs via SIRT4-Mediated Inactivation of JAK2/STAT3 Pathway. Int Heart J 2024; 65:738-747. [PMID: 39085113 DOI: 10.1536/ihj.23-631] [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] [Indexed: 08/02/2024]
Abstract
The regulatory factor X7 (RFX7) is a vital mediator in atherosclerosis. This study aims to discuss the effect and underlying mechanism of RFX7 on the regulation of oxidized low-density lipoprotein (ox-LDL) -induced proliferation and migration of vascular smooth muscle cells (VSMCs).Ox-LDL was used to construct atherosclerosis in vitro model. The mRNA and protein levels of RFX7 and Sirtuin 4 (SIRT4) were evaluated by quantitative real-time polymerase chain reaction (qRT-PCR) or western blot assays. The cellular functions were measured via 3-[4,5-dimethylthiazol-2-yl]-2,5-diphenyl tetrazolium bromide (MTT), EdU, flow cytometry, and wound healing assay assays. The interaction between RFX7 and SIRT4 promoter was validated using chromatin immunoprecipitation and dual-luciferase reporter assays.The stimulation with ox-LDL elevated the viability of VSMCs and decreased the mRNA and protein levels of RFX7 and SIRT4 in VSMCs in a dose-dependent manner. Functionally, RFX7 overexpression restrained the VSMC viability, proliferation, and migration induced by ox-LDL, but facilitated VSMC apoptosis. RFX7 elevated SIRT4 expression via binding to its promoter. Furthermore, overexpressing either SIRT4 or RFX7 inactivated JAK2/STAT3 signaling, causing a decrease in VSMC proliferation and migration and an increase in VSMC apoptosis when exposed to ox-LDL. The impact of RFX7 overexpression on JAK2/STAT3 signaling and cellular function following ox-LDL exposure was abrogated by SIRT4 silencing.The heightened RFX7 expression restrained the proliferation and migration of ox-LDL-stimulated VSMCs via SIRT4-mediated inactivation of JAK2/STAT3 pathway.
Collapse
Affiliation(s)
- Yinheng Hao
- Department of Interventional and Vascular Surgery, Suzhou Kowloon Hospital, Shanghai Jiao Tong University School of Medicine
| | - Wei Li
- Department of Vascular Surgery, The Second Hospital of Yinzhou District
| |
Collapse
|
6
|
Luo Y, Wang H, Wang L, Wu W, Zhao J, Li X, Xiong R, Ding X, Yuan D, Yuan C. LncRNA MEG3: Targeting the Molecular Mechanisms and Pathogenic causes of Metabolic Diseases. Curr Med Chem 2024; 31:6140-6153. [PMID: 37855346 DOI: 10.2174/0109298673268051231009075027] [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: 07/03/2023] [Revised: 08/27/2023] [Accepted: 09/08/2023] [Indexed: 10/20/2023]
Abstract
BACKGROUND Non-coding RNA is a type of RNA that does not encode proteins, distributed among rRNA, tRNA, snRNA, snoRNA, microRNA and other RNAs with identified functions, where the Long non-coding RNA (lncRNA) displays a nucleotide length over 200. LncRNAs enable multiple biological processes in the human body, including cancer cell invasion and metastasis, apoptosis, cell autophagy, inflammation, etc. Recently, a growing body of studies has demonstrated the association of lncRNAs with obesity and obesity-induced insulin resistance and NAFLD, where MEG3 is related to glucose metabolism, such as insulin resistance. In addition, MEG3 has been demonstrated in the pathological processes of various cancers, such as mediating inflammation, cardiovascular disease, liver disease and other metabolic diseases. OBJECTIVE To explore the regulatory role of lncRNA MEG3 in metabolic diseases. It provides new ideas for clinical treatment or experimental research. METHODS In this paper, in order to obtain enough data, we integrate and analyze the data in the PubMed database. RESULTS LncRNA MEG3 can regulate many metabolic diseases, such as insulin resistance, NAFLD, inflammation and so on. CONCLUSION LncRNA MEG3 has a regulatory role in a variety of metabolic diseases, which are currently difficult to be completely cured, and MEG3 is a potential target for the treatment of these diseases. Here, we review the role of lncRNA MEG3 in mechanisms of action and biological functions in human metabolic diseases.
Collapse
Affiliation(s)
- Yiyang Luo
- Third-grade Pharmacological Laboratory on Traditional Chinese Medicine, State Administration of Traditional Chinese Medicine, China Three Gorges University, Yichang, 443002, China
- College of Medicine and Health Science, China Three Gorges University, Yichang, 443002, China
| | - Hailin Wang
- Third-grade Pharmacological Laboratory on Traditional Chinese Medicine, State Administration of Traditional Chinese Medicine, China Three Gorges University, Yichang, 443002, China
- College of Medicine and Health Science, China Three Gorges University, Yichang, 443002, China
| | - Lijun Wang
- Third-grade Pharmacological Laboratory on Traditional Chinese Medicine, State Administration of Traditional Chinese Medicine, China Three Gorges University, Yichang, 443002, China
- Department of Biochemistry, College of Basic Medical Science, China Three Gorges University, Yichang, 443002, China
| | - Wei Wu
- Third-grade Pharmacological Laboratory on Traditional Chinese Medicine, State Administration of Traditional Chinese Medicine, China Three Gorges University, Yichang, 443002, China
- College of Medicine and Health Science, China Three Gorges University, Yichang, 443002, China
| | - Jiale Zhao
- Third-grade Pharmacological Laboratory on Traditional Chinese Medicine, State Administration of Traditional Chinese Medicine, China Three Gorges University, Yichang, 443002, China
- College of Medicine and Health Science, China Three Gorges University, Yichang, 443002, China
| | - Xueqing Li
- Third-grade Pharmacological Laboratory on Traditional Chinese Medicine, State Administration of Traditional Chinese Medicine, China Three Gorges University, Yichang, 443002, China
- College of Medicine and Health Science, China Three Gorges University, Yichang, 443002, China
| | - Ruisi Xiong
- Third-grade Pharmacological Laboratory on Traditional Chinese Medicine, State Administration of Traditional Chinese Medicine, China Three Gorges University, Yichang, 443002, China
- Department of Biochemistry, College of Basic Medical Science, China Three Gorges University, Yichang, 443002, China
| | - Xueliang Ding
- Department of Clinical Laboratory, Affiliated Renhe Hospital of China Three Gorges University, Yichang, 443002, China
| | - Ding Yuan
- Third-grade Pharmacological Laboratory on Traditional Chinese Medicine, State Administration of Traditional Chinese Medicine, China Three Gorges University, Yichang, 443002, China
- College of Medicine and Health Science, China Three Gorges University, Yichang, 443002, China
| | - Chengfu Yuan
- Third-grade Pharmacological Laboratory on Traditional Chinese Medicine, State Administration of Traditional Chinese Medicine, China Three Gorges University, Yichang, 443002, China
- Department of Biochemistry, College of Basic Medical Science, China Three Gorges University, Yichang, 443002, China
| |
Collapse
|
7
|
Zheng Y, Wang S, Wu J, Wang Y. Mitochondrial metabolic dysfunction and non-alcoholic fatty liver disease: new insights from pathogenic mechanisms to clinically targeted therapy. J Transl Med 2023; 21:510. [PMID: 37507803 PMCID: PMC10375703 DOI: 10.1186/s12967-023-04367-1] [Citation(s) in RCA: 38] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Accepted: 07/17/2023] [Indexed: 07/30/2023] Open
Abstract
Metabolic dysfunction-associated fatty liver disease (MAFLD) is among the most widespread metabolic disease globally, and its associated complications including insulin resistance and diabetes have become threatening conditions for human health. Previous studies on non-alcoholic fatty liver disease (NAFLD) were focused on the liver's lipid metabolism. However, growing evidence suggests that mitochondrial metabolism is involved in the pathogenesis of NAFLD to varying degrees in several ways, for instance in cellular division, oxidative stress, autophagy, and mitochondrial quality control. Ultimately, liver function gradually declines as a result of mitochondrial dysfunction. The liver is unable to transfer the excess lipid droplets outside the liver. Therefore, how to regulate hepatic mitochondrial function to treat NAFLD has become the focus of current research. This review provides details about the intrinsic link of NAFLD with mitochondrial metabolism and the mechanisms by which mitochondrial dysfunctions contribute to NAFLD progression. Given the crucial role of mitochondrial metabolism in NAFLD progression, the application potential of multiple mitochondrial function improvement modalities (including physical exercise, diabetic medications, small molecule agonists targeting Sirt3, and mitochondria-specific antioxidants) in the treatment of NAFLD was evaluated hoping to provide new insights into NAFLD treatment.
Collapse
Affiliation(s)
- Youwei Zheng
- Department of General Surgery, The Fourth Affiliated Hospital of China Medical University, Shenyang, Liaoning Province, China
| | - Shiting Wang
- Department of Cardiovascular Medicine, The Fourth Affiliated Hospital of China Medical University, Shenyang, China
| | - Jialiang Wu
- Department of General Surgery, The Fourth Affiliated Hospital of China Medical University, Shenyang, Liaoning Province, China
| | - Yong Wang
- Department of General Surgery, The Fourth Affiliated Hospital of China Medical University, Shenyang, Liaoning Province, China.
| |
Collapse
|
8
|
Chang S, Zhang G, Li L, Li H, Jin X, Wang Y, Li B. Sirt4 deficiency promotes the development of atherosclerosis by activating the NF-κB/IκB/CXCL2/3 pathway. Atherosclerosis 2023; 373:29-37. [PMID: 37121164 DOI: 10.1016/j.atherosclerosis.2023.04.006] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/06/2022] [Revised: 04/06/2023] [Accepted: 04/20/2023] [Indexed: 05/02/2023]
Abstract
BACKGROUND AND AIMS As a member of mitochondrial sirtuins, Sirt4 plays a vital role in cellular metabolism and intracellular signal transduction; however, its effect on atherosclerosis is unclear. This study aimed to explore the effect of Sirt4 on atherosclerosis and its underlying mechanism. METHODS In vivo, Apoe-/- and Apoe-/-/Sirt4-/- mice were fed a high-fat diet to induce atherosclerosis. In vitro, peritoneal macrophages from two mouse types were extracted and treated with oxidized low-density lipoprotein to establish a cell model, THP-1 cells were used to observe the effect of Sirt4 on the adhesion ability of monocytes. The growth and composition of aortic plaques in two mouse types were analyzed by H&E staining, Oil Red O staining, Dil oxidized low-density lipoprotein, immunohistochemistry, real-time quantitative polymerase chain reaction and enzyme-linked immunosorbent assay. Transcriptome analysis and Western blotting were performed to explore the specific mechanism. RESULTS Sirt4 deficiency aggravated atherosclerosis in mice. In vivo, aortic plaque size, lipid content, and expression of related inflammatory factors in Apoe-/-/Sirt4-/- mice were higher than those in the control group, whereas the content of collagen Ⅰ and smooth muscle actin-α was significantly lower. Sirt4-deficient macrophages exhibited stronger lipid phagocytosis in vitro, and the adhesion ability of monocytes increased when Sirt4 expression decreased. Transcriptome analysis showed that the expression of CXCL2 and CXCL3 in Sirt4-deficient peritoneal macrophages increased significantly, which may play a role by activating the NF-κB pathway. In further analysis, the results in vitro and in vivo showed that the expression of VCAM-1 and pro-inflammatory factors, such as IL-6, TNF-α and IL-1β, increased, whereas the expression of anti-inflammatory factor IL-37 decreased in Sirt4-deficient peritoneal macrophages and tissues. After blocking the effect with NK-κB inhibitor BAY11-7082, the inflammatory reaction in sirt4 deficient macrophages was also significantly decreased. CONCLUSIONS This study demonstrates that Sirt4 deficiency promotes the development of atherosclerosis by activating the NF-κB/IκB/CXCL2/3 pathway, suggesting that Sirt4 may exhibit a protective effect in atherosclerosis, which provides a new strategy for clinical prevention and treatment of atherosclerosis.
Collapse
Affiliation(s)
- Shuting Chang
- Department of Cardiology, Zibo Central Hospital Affiliated to Binzhou Medical College, NO.10, South Shanghai Road, Zibo, PR China; Weifang Medical University, No.7166, Baotong West Street, Weifang, PR China
| | - Guanzhao Zhang
- Department of Cardiology, Zibo Central Hospital Affiliated to Binzhou Medical College, NO.10, South Shanghai Road, Zibo, PR China
| | - Lanlan Li
- Center of Translational Medicine, Zibo Central Hospital, No. 10, South Shanghai Road, Zibo, PR China
| | - Haiying Li
- Medical Department, Zibo Central Hospital, No. 10, South Shanghai Road, Zibo, PR China
| | - Xiaodong Jin
- Department of Geriatrics, Zibo Central Hospital, No. 10, South Shanghai Road, Zibo, PR China
| | - Yunshan Wang
- Department of Clinical Laboratory, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, PR China.
| | - Bo Li
- Department of Cardiology, Zibo Central Hospital Affiliated to Binzhou Medical College, NO.10, South Shanghai Road, Zibo, PR China.
| |
Collapse
|
9
|
He L, Liu Q, Cheng J, Cao M, Zhang S, Wan X, Li J, Tu H. SIRT4 in ageing. Biogerontology 2023; 24:347-362. [PMID: 37067687 DOI: 10.1007/s10522-023-10022-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2022] [Accepted: 01/31/2023] [Indexed: 04/18/2023]
Abstract
Ageing is a phenomenon in which cells, tissues and organs undergo systemic pathological changes as individuals age, leading to the occurrence of ageing-related diseases and the end of life. It is associated with many phenotypes known as ageing characteristics, such as genomic instability, nutritional imbalance, mitochondrial dysfunction, cell senescence, stem cell depletion, and an altered microenvironment. The sirtuin family (SIRT), known as longevity proteins, is thought to delay ageing and prolong life, and mammals, including humans, have seven family members (SIRT1-7). SIRT4 has been studied less among the sirtuin family thus far, but it has been reported that it has important physiological functions in organisms, such as promoting DNA damage repair, participating in the energy metabolism of three substances, inhibiting inflammatory reactions and apoptosis, and regulating mitochondrial function. Recently, some studies have demonstrated the involvement of SIRT4 in age-related processes, but knowledge in this field is still scarce. Therefore, this review aims to analyse the relationship between SIRT4 and ageing characteristics as well as some age-related diseases (e.g., cardiovascular diseases, metabolic diseases, neurodegenerative diseases and cancer).
Collapse
Affiliation(s)
- Ling He
- The Department of Geratology, The Second Affiliated Hospital of Nanchang University, 1 Minde Road, Nanchang, 330006, Jiangxi, China
| | - Qingcheng Liu
- The Department of Geratology, The Second Affiliated Hospital of Nanchang University, 1 Minde Road, Nanchang, 330006, Jiangxi, China
| | - Jielong Cheng
- The Department of Geratology, The Second Affiliated Hospital of Nanchang University, 1 Minde Road, Nanchang, 330006, Jiangxi, China
| | - Mei Cao
- The Department of Geratology, The Second Affiliated Hospital of Nanchang University, 1 Minde Road, Nanchang, 330006, Jiangxi, China
| | - Shuaimei Zhang
- The Department of Geratology, The Second Affiliated Hospital of Nanchang University, 1 Minde Road, Nanchang, 330006, Jiangxi, China
| | - Xiaolin Wan
- The Department of Geratology, The Second Affiliated Hospital of Nanchang University, 1 Minde Road, Nanchang, 330006, Jiangxi, China
| | - Jian Li
- The Key Laboratory of Hematology of Jiangxi Province, The Second Affiliated Hospital of Nanchang University, 1 Minde Road, Nanchang, 330006, Jiangxi, China.
| | - Huaijun Tu
- The Department of Geratology, The Second Affiliated Hospital of Nanchang University, 1 Minde Road, Nanchang, 330006, Jiangxi, China.
| |
Collapse
|
10
|
Luo Y, Jiao Q, Chen Y. Targeting endoplasmic reticulum stress-the responder to lipotoxicity and modulator of non-alcoholic fatty liver diseases. Expert Opin Ther Targets 2022; 26:1073-1085. [PMID: 36657744 DOI: 10.1080/14728222.2022.2170780] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
INTRODUCTION Endoplasmic reticulum (ER) stress occurs with aberrant lipid accumulation and resultant adverse effects and widely exists in nonalcoholic fatty liver disease (NAFLD). It triggers the unfolded protein response (UPR) to restore ER homeostasis and actively participates in NAFLD pathological processes, including hepatic steatosis, inflammation, hepatocyte death, and fibrosis. Such acknowledges drive the discovery of novel NAFLD biomarker and therapeutic targets and the development of ER-stress targeted NAFLD drugs. AREAS COVERED This article discusses and updates the role of ER stress and UPR in NAFLD, the underlying action mechanism, and especially their full participation in NAFLD pathophysiology. It characterizes key molecular targets useful for the prevention and treatment of NAFLD and highlights the recent ER stress-targeted therapeutic strategies for NAFLD. EXPERT OPINION Targeting ER Stress is a valuable and promising strategy for NAFLD treatment, but its smooth translation into clinical application still requires better clarification of the different UPR patterns in diverse NAFLD physiological states. Further understanding of the distinct effects of these various patterns on NAFLD, the thresholds deciding their final impacts, and their actions via non-liver tissues and cells would be of great help to develop a precise and effective therapy for NAFLD. [Figure: see text].
Collapse
Affiliation(s)
- Yu Luo
- School of Pharmaceutical Science, University of South China, Hengyang, Hunan, China
| | - Qiangqiang Jiao
- School of Pharmaceutical Science, University of South China, Hengyang, Hunan, China
| | - Yuping Chen
- School of Pharmaceutical Science, University of South China, Hengyang, Hunan, China.,Institute of Pharmacy & Pharmacology, University of South China, Hengyang, Hunan, China
| |
Collapse
|
11
|
Wimalarathne MM, Wilkerson-Vidal QC, Hunt EC, Love-Rutledge ST. The case for FAT10 as a novel target in fatty liver diseases. Front Pharmacol 2022; 13:972320. [PMID: 36386217 PMCID: PMC9665838 DOI: 10.3389/fphar.2022.972320] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2022] [Accepted: 10/12/2022] [Indexed: 12/13/2022] Open
Abstract
Human leukocyte antigen F locus adjacent transcript 10 (FAT10) is a ubiquitin-like protein that targets proteins for degradation. TNFα and IFNγ upregulate FAT10, which increases susceptibility to inflammation-driven diseases like nonalcoholic fatty liver disease (NAFLD), non-alcoholic steatohepatitis (NASH), and hepatocellular carcinoma (HCC). It is well established that inflammation contributes to fatty liver disease, but how inflammation contributes to upregulation and what genes are involved is still poorly understood. New evidence shows that FAT10 plays a role in mitophagy, autophagy, insulin signaling, insulin resistance, and inflammation which may be directly associated with fatty liver disease development. This review will summarize the current literature regarding FAT10 role in developing liver diseases and potential therapeutic targets for nonalcoholic/alcoholic fatty liver disease and hepatocellular carcinoma.
Collapse
|
12
|
Chen Y, Yang F, Chu Y, Yun Z, Yan Y, Jin J. Mitochondrial transplantation: opportunities and challenges in the treatment of obesity, diabetes, and nonalcoholic fatty liver disease. Lab Invest 2022; 20:483. [PMID: 36273156 PMCID: PMC9588235 DOI: 10.1186/s12967-022-03693-0] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Accepted: 10/06/2022] [Indexed: 11/23/2022]
Abstract
Metabolic diseases, including obesity, diabetes, and nonalcoholic fatty liver disease (NAFLD), are rising in both incidence and prevalence and remain a major global health and socioeconomic burden in the twenty-first century. Despite an increasing understanding of these diseases, the lack of effective treatments remains an ongoing challenge. Mitochondria are key players in intracellular energy production, calcium homeostasis, signaling, and apoptosis. Emerging evidence shows that mitochondrial dysfunction participates in the pathogeneses of metabolic diseases. Exogenous supplementation with healthy mitochondria is emerging as a promising therapeutic approach to treating these diseases. This article reviews recent advances in the use of mitochondrial transplantation therapy (MRT) in such treatment.
Collapse
Affiliation(s)
- Yifei Chen
- Department of Laboratory Medicine, Wujin Hospital Affiliated With Jiangsu University (The Wujin Clinical College of Xuzhou Medical University), Changzhou, 213017, Jiangsu Province, China.,School of Medicine, Jiangsu University, ZhenjiangJiangsu Province, 212013, China
| | - Fuji Yang
- Department of Laboratory Medicine, Wujin Hospital Affiliated With Jiangsu University (The Wujin Clinical College of Xuzhou Medical University), Changzhou, 213017, Jiangsu Province, China.,School of Medicine, Jiangsu University, ZhenjiangJiangsu Province, 212013, China
| | - Ying Chu
- Department of Laboratory Medicine, Wujin Hospital Affiliated With Jiangsu University (The Wujin Clinical College of Xuzhou Medical University), Changzhou, 213017, Jiangsu Province, China.,Central Laboratory, Wujin Hospital Affiliated With Jiangsu University (The Wujin Clinical College of Xuzhou Medical University), Changzhou, 213017, Jiangsu Province, China
| | - Zhihua Yun
- Department of Laboratory Medicine, Wujin Hospital Affiliated With Jiangsu University (The Wujin Clinical College of Xuzhou Medical University), Changzhou, 213017, Jiangsu Province, China
| | - Yongmin Yan
- Department of Laboratory Medicine, Wujin Hospital Affiliated With Jiangsu University (The Wujin Clinical College of Xuzhou Medical University), Changzhou, 213017, Jiangsu Province, China. .,Central Laboratory, Wujin Hospital Affiliated With Jiangsu University (The Wujin Clinical College of Xuzhou Medical University), Changzhou, 213017, Jiangsu Province, China.
| | - Jianhua Jin
- Department of Oncology, Wujin Hospital Affiliated With Jiangsu University (The Wujin Clinical College of Xuzhou Medical University), Changzhou, 213017, Jiangsu Province, China.
| |
Collapse
|
13
|
Sodium butyrate reduces overnutrition-induced microglial activation and hypothalamic inflammation. Int Immunopharmacol 2022; 111:109083. [PMID: 35917736 DOI: 10.1016/j.intimp.2022.109083] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Revised: 07/13/2022] [Accepted: 07/20/2022] [Indexed: 11/21/2022]
|
14
|
Yang B, Lu L, Zhou D, Fan W, Barbier-Torres L, Steggerda J, Yang H, Yang X. Regulatory network and interplay of hepatokines, stellakines, myokines and adipokines in nonalcoholic fatty liver diseases and nonalcoholic steatohepatitis. Front Endocrinol (Lausanne) 2022; 13:1007944. [PMID: 36267567 PMCID: PMC9578007 DOI: 10.3389/fendo.2022.1007944] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Accepted: 09/05/2022] [Indexed: 11/29/2022] Open
Abstract
Fatty liver disease is a spectrum of liver pathologies ranging from simple hepatic steatosis to non-alcoholic fatty liver disease (NAFLD), non-alcoholic steatohepatitis (NASH), and culminating with the development of cirrhosis or hepatocellular carcinoma (HCC). The pathogenesis of NAFLD is complex and diverse, and there is a lack of effective treatment measures. In this review, we address hepatokines identified in the pathogenesis of NAFLD and NASH, including the signaling of FXR/RXR, PPARα/RXRα, adipogenesis, hepatic stellate cell activation/liver fibrosis, AMPK/NF-κB, and type 2 diabetes. We also highlight the interaction between hepatokines, and cytokines or peptides secreted from muscle (myokines), adipose tissue (adipokines), and hepatic stellate cells (stellakines) in response to certain nutritional and physical activity. Cytokines exert autocrine, paracrine, or endocrine effects on the pathogenesis of NAFLD and NASH. Characterizing signaling pathways and crosstalk amongst muscle, adipose tissue, hepatic stellate cells and other liver cells will enhance our understanding of interorgan communication and potentially serve to accelerate the development of treatments for NAFLD and NASH.
Collapse
Affiliation(s)
- Bing Yang
- Department of Geriatric Endocrinology and Metabolism, Guangxi Key Laboratory of Precision Medicine in Cardio-cerebrovascular Diseases Control and Prevention, Guangxi Clinical Research Center for Cardio-cerebrovascular Diseases, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Liqing Lu
- Department of Thoracic Surgery, Xiangya Hospital, Central South University, Changsha, China
| | - Dongmei Zhou
- Department of Geriatric Endocrinology and Metabolism, Guangxi Key Laboratory of Precision Medicine in Cardio-cerebrovascular Diseases Control and Prevention, Guangxi Clinical Research Center for Cardio-cerebrovascular Diseases, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Wei Fan
- Division of Digestive and Liver Diseases, Cedars-Sinai Medical Center, Los Angeles, CA, United States
| | - Lucía Barbier-Torres
- Division of Digestive and Liver Diseases, Cedars-Sinai Medical Center, Los Angeles, CA, United States
| | - Justin Steggerda
- Department of Surgery, Cedars-Sinai Medical Center, Los Angeles, CA, United States
| | - Heping Yang
- Division of Digestive and Liver Diseases, Cedars-Sinai Medical Center, Los Angeles, CA, United States
| | - Xi Yang
- Department of Geriatric Endocrinology and Metabolism, Guangxi Key Laboratory of Precision Medicine in Cardio-cerebrovascular Diseases Control and Prevention, Guangxi Clinical Research Center for Cardio-cerebrovascular Diseases, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| |
Collapse
|
15
|
Ding W, Cai C, Zhu X, Wang J, Jiang Q. Parthenolide ameliorates neurological deficits and neuroinflammation in mice with traumatic brain injury by suppressing STAT3/NF-κB and inflammasome activation. Int Immunopharmacol 2022; 108:108913. [PMID: 35729839 DOI: 10.1016/j.intimp.2022.108913] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Revised: 05/15/2022] [Accepted: 05/27/2022] [Indexed: 01/10/2023]
Abstract
BACKGROUND Traumatic brain injury (TBI) triggers a set of complex inflammation that results in secondary injury. Parthenolide (PTN) is a sesquiterpene lactone extracted from the herb Tanacetum parthenium (Feverfew) and has potent anti-inflammatory, anti-apoptosis and anti-oxidative stress effects in the central nervous system (CNS)-related diseases. This study focuses on investigating the potential neuroprotective effect of PTN on TBI and the related mechanism. METHODS Bv2 microglia, primary microglia were stimulated by LPS, and HT22 neuron cells were stimulated by OGD/R, and they were treated with different doses of PTN. The expression profiles of pro-inflammatory cytokines, proteins, oxidative stress mediators, STAT3/NF-κB pathway, inflammasomes were detected. Forty male/female C57BL/6 mice were randomly divided into the sham, PTN, TBI, and TBI + PTN groups (10 mice per group). A mouse TBI model was set up with a controlled cortical impact (CCI) device. The modified nerve severity score (mNSS) was implemented to check short-term neurological impairment in mice, and the mice's memory and learning were assessed by the Morris water maze test. The water content in the mice's brains was measured by the dry-wet method. Hematoxylin-eosin (H&E) staining, Nissl staining and terminal deoxynucleotidyl transferase-mediated dUTP-biotin nick end labeling (TUNEL) assay were applied for neuronal apoptosis. RESULTS PTN dramatically alleviated LPS-induced inflammation in microglia, and OGD-mediated neuronal apoptosis and oxidative stress. In addition, PTN repressed LPS- or OGD-modulated STAT3/NF-κB and NLR family pyrin domain containing 1 (NLRP1), NLRP3, NLR family CARD domain containing 4 (NLRC4) inflammasomes activation. Administering the STAT3 inhibitor Stattic or NF-κB inhibitor Bay 11-7082 attenuated PTN-mediated effects. In vivo, PTN treatment relieved neural function deficits, brain edema and neuron apoptosis and improved the memory and learning function of TBI mice. Additionally, PTN impeded microglial activation and reduced the production of pro-inflammatory cytokines in brain lesions of TBI mice. Furthermore, PTN hindered STAT3/NF-κB and inflammasome activation. CONCLUSION PTN can curb microglial activation and neuron apoptosis by dampening the STAT3/NF-κB pathway, thus exerting neuroprotective effects in TBI mice.
Collapse
Affiliation(s)
- Wei Ding
- Department of Neurosurgery, Tianyou Hospital Affiliated to Wuhan University of Science and Technology, Wuhan 430000, China; Department of Neurosurgery, Liyuan Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China.
| | - Chen Cai
- State Key Laboratory of Agricultural Microbiology, College of Animal Science and Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China
| | - Xiaomin Zhu
- Department of Neurology, Guangxi University of Chinese Medicine, Nanning 530200 Guangxi, China
| | - Jing Wang
- State Key Laboratory of Agricultural Microbiology, College of Animal Science and Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China
| | - Qian Jiang
- Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong, University of Science and Technology, Wuhan 430030, China.
| |
Collapse
|
16
|
Meng Q, Li X, Xiong X. Identification of Hub Genes Associated With Non-alcoholic Steatohepatitis Using Integrated Bioinformatics Analysis. Front Genet 2022; 13:872518. [PMID: 35559030 PMCID: PMC9086399 DOI: 10.3389/fgene.2022.872518] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Accepted: 03/31/2022] [Indexed: 12/30/2022] Open
Abstract
Background and aims: As a major cause of liver disease worldwide, non-alcoholic fatty liver disease (NAFLD) comprises non-alcoholic fatty liver (NAFL) and non-alcoholic steatohepatitis (NASH). Due to the high prevalence and poor prognosis of NASH, it is critical to understand its mechanisms. However, the etiology and mechanisms remain largely unknown. In addition, the gold standard for the diagnosis of NASH is liver biopsy, which is an invasive procedure. Therefore, there is a pressing need to develop noninvasive tests for NASH diagnosis. The goal of the study is to discover key genes involved in NASH development and investigate their value as noninvasive biomarkers. Methods: The Gene Expression Omnibus (GEO) database was used to obtain two datasets encompassing NASH patients and healthy controls. We used weighted gene co-expression network analysis (WGCNA) and differential expression analysis in order to investigate the association between gene sets and clinical features, as well as to discover co-expression modules. A protein-protein interaction (PPI) network was created to extract hub genes. The results were validated using another publicly available dataset and mice treated with a high-fat diet (HFD) and carbon tetrachloride (CCl4). Results: A total of 24 differentially co-expressed genes were selected by WGCNA and differential expression analysis. KEGG analysis indicated most of them were enriched in the focal adhesion pathway. GO analysis showed these genes were mainly enriched in circadian rhythm, aging, angiogenesis and response to drug (biological process), endoplasmic reticulum lumen (cellular component), and protein binding (molecular function). As a result, eight genes (JUN, SERPINE1, GINS2, TYMS, HMMR, IGFBP2, BIRC3, TNFRSF12A) were identified as hub genes. Finally, three genes were found significantly changed in both the validation dataset and the mouse model. Conclusion: Our research discovered genes that have the potential to mediate the process of NASH and might be useful diagnostic biomarkers for the disorder.
Collapse
Affiliation(s)
- Qingnan Meng
- The Key Laboratory of Metabolism and Molecular Medicine of the Ministry of Education, Department of Endocrinology and Metabolism, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Xiaoying Li
- The Key Laboratory of Metabolism and Molecular Medicine of the Ministry of Education, Department of Endocrinology and Metabolism, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Xuelian Xiong
- The Key Laboratory of Metabolism and Molecular Medicine of the Ministry of Education, Department of Endocrinology and Metabolism, Zhongshan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
17
|
Huang T, Gao Y, Cao Y, Wang Q, Dong Z. Downregulation of mmu_circ_0000943 ameliorates renal ischemia reperfusion-triggered inflammation and oxidative stress via regulating mmu-miR-377-3p/Egr2 axis. Int Immunopharmacol 2022; 106:108614. [PMID: 35168080 DOI: 10.1016/j.intimp.2022.108614] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Revised: 01/29/2022] [Accepted: 02/05/2022] [Indexed: 12/31/2022]
|
18
|
Abstract
Non-alcoholic fatty liver disease (NAFLD) is a common disease defined by excess fat deposition in the liver. The course of NAFLD is not fully understood, however, some pathogenic mechanisms have been identified. Accumulation of fat in liver cells is associated with insulin resistance, central obesity, triglyceride accumulation in the liver and hepatic fatty acid metabolism dysregulation that cause steatosis. The other process leads to hepatocyte inflammation and necrosis, which leads to severe hepatic disease; non-alcoholic steatohepatitis. Many clinical studies have underlined the link between NAFLD and atherosclerosis. NAFLD may alter the balance lipid-glucose metabolism as well as increase the risk of hypertension and systemic inflammation. This results in a greater risk of vascular events. The present review considers the link between NAFLD and atherosclerosis.
Collapse
Affiliation(s)
- Sevket Balta
- Department of Cardiology, Hayat Hospital, Malatya, Turkey
| |
Collapse
|
19
|
Li N, Tan H, Xie A, Li C, Fu X, Xang W, Kirim A, Huang X. Value of the triglyceride glucose index combined with body mass index in identifying non-alcoholic fatty liver disease in patients with type 2 diabetes. BMC Endocr Disord 2022; 22:101. [PMID: 35428219 PMCID: PMC9011983 DOI: 10.1186/s12902-022-00993-w] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Accepted: 03/16/2022] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND The triglyceride glucose index combined with body mass index is a new index that reflects the degree of insulin resistance. In this cross-sectional study, we aimed to explore the predictive value of the triglyceride glucose-body mass index (TyG-BMI) in relation to the occurrence of non-alcoholic fatty liver disease (NAFLD) in the Chinese population with type 2 diabetes (T2D). METHODS We selected 826 patients with T2D who were hospitalized at the Department of Endocrinology and Metabolism of Karamay People's Hospital from September 2016 to October 2018 for this research. The height, weight, fasting blood glucose, serum insulin, and lipid profiles of the subjects were collected. The liver ultrasound showed any degree of echogenic enhancement of liver tissue and the liver appeared brighter than the renal cortex on ultrasound were considered to be NAFLD. The logistic regression analysis was performed to estimate associations between the triglyceride glucose index (TyG), TyG-BMI index, insulin resistance index (HOMA-IR) and the ratio of the triglycerides to high-density lipoprotein-cholesterol with a diagnosis of NAFLD. The receiver operating characteristic curve method was used to analyze its predictive value for NAFLD. RESULTS Results of the logistic regression analysis showed that the odds ratios of NAFLD were 6.535 (3.70-11.53) and 4.868 (2.576-9.200) for the TyG-BMI before and after correction,respectively(P < 0.001). The area under the curve (AUC) for TyG-BMI was 0.727 (0.691-0.764), which was the highest among all the other parameters studied. CONCLUSION Compared with the TyG index, the TG/HDL-C and HOMA-IR, the TyG-BMI was a more effective predictor of NAFLD in T2D.
Collapse
Affiliation(s)
- Nong Li
- Department of Endocrinology and Metabolism, People's Hospital of Karamay, Xinjiang, China.
| | - Huiwen Tan
- Department of Endocrinology Metabolism, West China Hospital of Sichuan University, Chengdu, China
| | - Aixia Xie
- Department of Endocrinology and Metabolism, People's Hospital of Karamay, Xinjiang, China
| | - Cheng Li
- First Affiliated Hospital of Xinjiang Medical University, Ürümqi, China
| | - Xuan Fu
- Department of Endocrinology and Metabolism, People's Hospital of Karamay, Xinjiang, China
| | - Weiting Xang
- Department of Endocrinology and Metabolism, People's Hospital of Karamay, Xinjiang, China
| | - Amina Kirim
- Department of Endocrinology and Metabolism, People's Hospital of Karamay, Xinjiang, China
| | - Xuefang Huang
- Department of Endocrinology and Metabolism, People's Hospital of Karamay, Xinjiang, China.
| |
Collapse
|
20
|
Sun CY, Zheng ZL, Chen CW, Lu BW, Liu D. Targeting Gut Microbiota With Natural Polysaccharides: Effective Interventions Against High-Fat Diet-Induced Metabolic Diseases. Front Microbiol 2022; 13:859206. [PMID: 35369480 PMCID: PMC8965082 DOI: 10.3389/fmicb.2022.859206] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Accepted: 02/22/2022] [Indexed: 12/12/2022] Open
Abstract
Unhealthy diet, in particular high-fat diet (HFD) intake, can cause the development of several metabolic disorders, including obesity, hyperlipidemia, type 2 diabetes mellitus (T2DM), non-alcoholic fatty liver disease (NAFLD), and metabolic syndrome (MetS). These popular metabolic diseases reduce the quality of life, and induce premature death worldwide. Evidence is accumulating that the gut microbiota is inextricably associated with HFD-induced metabolic disorders, and dietary intervention of gut microbiota is an effective therapeutic strategy for these metabolic dysfunctions. Polysaccharides are polymeric carbohydrate macromolecules and sources of fermentable dietary fiber that exhibit biological activities in the prevention and treatment of HFD-induced metabolic diseases. Of note, natural polysaccharides are among the most potent modulators of the gut microbiota composition. However, the prebiotics-like effects of polysaccharides in treating HFD-induced metabolic diseases remain elusive. In this review, we introduce the critical role of gut microbiota human health and HFD-induced metabolic disorders. Importantly, we review current knowledge about the role of natural polysaccharides in improving HFD-induced metabolic diseases by regulating gut microbiota.
Collapse
Affiliation(s)
- Chao-Yue Sun
- College of Biological and Pharmaceutical Engineering, West Anhui University, Lu'an, China
| | | | - Cun-Wu Chen
- College of Biological and Pharmaceutical Engineering, West Anhui University, Lu'an, China
| | - Bao-Wei Lu
- College of Biological and Pharmaceutical Engineering, West Anhui University, Lu'an, China
| | - Dong Liu
- College of Biological and Pharmaceutical Engineering, West Anhui University, Lu'an, China
| |
Collapse
|
21
|
Wang Y, Wang Y, Li F, Zou J, Li X, Xu M, Yu D, Ma Y, Huang W, Sun X, Zhang Y. Psoralen Suppresses Lipid Deposition by Alleviating Insulin Resistance and Promoting Autophagy in Oleate-Induced L02 Cells. Cells 2022; 11:cells11071067. [PMID: 35406631 PMCID: PMC8997557 DOI: 10.3390/cells11071067] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2022] [Revised: 03/09/2022] [Accepted: 03/17/2022] [Indexed: 11/16/2022] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) held a high global prevalence in recent decades. Hepatic lipid deposition is the major characteristic of NAFLD. We aim to explore the mechanisms of psoralen on lipid deposition in NAFLD. The effects of psoralen on insulin resistance, lipid deposition, the expression and membrane translocation of glucose transporter type 4 (GLUT4), autophagy, and lipogenesis enzymes were determined on sodium oleate-induced L02 cells. Chloroquine and 3-MA were employed. The AMP-activated protein kinase alpha (AMPKα) was knocked down by siRNA. Psoralen alleviated insulin resistance in sodium oleate-induced L02 hepatocytes by upregulating the expression and membrane translocation of GLUT4. Psoralen inhibited lipid accumulation by decreasing the expression of key lipogenesis enzymes. Psoralen promotes autophagy and the autophagic flux to enhance lipolysis. Psoralen promoted the fusion of the autophagosome with the lysosome. Both chloroquine and 3-MA blocked the effects of psoralen on autophagy and lipid accumulation. The AMPKα deficiency attenuated the effects of psoralen on autophagy and lipid accumulation. Our study demonstrated that as an antioxidant, psoralen attenuates NAFLD by alleviating insulin resistance and promoting autophagy via AMPK, suggesting psoralen to be a promising candidate for NAFLD.
Collapse
Affiliation(s)
- Yuhao Wang
- West China School of Pharmacy, Sichuan University, Chengdu 610041, China;
- West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu 610041, China; (Y.W.); (J.Z.); (X.L.); (M.X.); (Y.M.)
| | - Yonglun Wang
- West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu 610041, China; (Y.W.); (J.Z.); (X.L.); (M.X.); (Y.M.)
| | - Fang Li
- Department of Hepatopancreatobiliary Surgery, Sichuan Cancer Hospital and Institute, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610041, China;
| | - Jie Zou
- West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu 610041, China; (Y.W.); (J.Z.); (X.L.); (M.X.); (Y.M.)
| | - Xiaoqian Li
- West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu 610041, China; (Y.W.); (J.Z.); (X.L.); (M.X.); (Y.M.)
| | - Mengxia Xu
- West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu 610041, China; (Y.W.); (J.Z.); (X.L.); (M.X.); (Y.M.)
| | - Daojiang Yu
- Department of Plastic Surgery, The Second Affiliated Hospital of Chengdu Medical College, China National Nuclear Corporation 416 Hospital, Chengdu 610051, China;
| | - Yijia Ma
- West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu 610041, China; (Y.W.); (J.Z.); (X.L.); (M.X.); (Y.M.)
| | - Wei Huang
- Department of Endocrinology, Affiliated Hospital of Southwest Medical University, Luzhou 646000, China;
| | - Xiaodong Sun
- West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu 610041, China; (Y.W.); (J.Z.); (X.L.); (M.X.); (Y.M.)
- Correspondence: (X.S.); or (Y.Z.); Tel.: +86-28-8550-1278 (X.S. & Y.Z.)
| | - Yuanyuan Zhang
- West China School of Pharmacy, Sichuan University, Chengdu 610041, China;
- West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu 610041, China; (Y.W.); (J.Z.); (X.L.); (M.X.); (Y.M.)
- Correspondence: (X.S.); or (Y.Z.); Tel.: +86-28-8550-1278 (X.S. & Y.Z.)
| |
Collapse
|
22
|
Yan B, Chen L, Wang Y, Zhang J, Zhao H, Hua Q, Pei S, Yue Z, Liang H, Zhang H. Preventive Effect of Apple Polyphenol Extract on High-Fat Diet-Induced Hepatic Steatosis in Mice through Alleviating Endoplasmic Reticulum Stress. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2022; 70:3172-3180. [PMID: 35227062 DOI: 10.1021/acs.jafc.1c07733] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
In this work, the protective effect of apple polyphenol extract (APE) on hepatic steatosis was investigated. Thirty-two C57BL/6J mice were assigned randomly to control group, hepatic steatosis group, lovastatin group, and APE group. After 8 weeks of intervention, APE supplementation markedly decreased the body weight gain, liver weight, liver index, epididymal adipose weight, epididymal adipose index, serum, and hepatic lipid levels. Hematoxylin and eosin staining revealed that APE supplementation alleviated histopathological changes of hepatic steatosis. Western blot revealed that APE downregulated the protein levels of GRP78, IRE1α, p-IRE1α, XBP1, PERK, p-PERK, p-eIF2α, ATF6, PPAR-γ, SREBP-1c, FAS, and ACC1. In conclusion, this study found that APE inhibited IRE1α-XBP1, PERK-eIF2α, and ATF6 signaling pathways to alleviate endoplasmic reticulum stress, thereby improving HFD-induced hepatic steatosis.
Collapse
Affiliation(s)
- Bei Yan
- Department of Nutrition and Food Hygiene, College of Public Health, Qingdao University, 308 Ningxia Road, Qingdao 266071, China
| | - Lei Chen
- Department of Nutrition and Food Hygiene, College of Public Health, Qingdao University, 308 Ningxia Road, Qingdao 266071, China
| | - Yanhui Wang
- Department of Nutrition and Food Hygiene, College of Public Health, Qingdao University, 308 Ningxia Road, Qingdao 266071, China
| | - Jiacheng Zhang
- Department of Nutrition and Food Hygiene, College of Public Health, Qingdao University, 308 Ningxia Road, Qingdao 266071, China
| | - Hui Zhao
- Department of Nutrition and Food Hygiene, College of Public Health, Qingdao University, 308 Ningxia Road, Qingdao 266071, China
| | - Qinglian Hua
- Department of Nutrition and Food Hygiene, College of Public Health, Qingdao University, 308 Ningxia Road, Qingdao 266071, China
| | - Shengjie Pei
- Department of Nutrition and Food Hygiene, College of Public Health, Qingdao University, 308 Ningxia Road, Qingdao 266071, China
| | - Zihang Yue
- Department of Nutrition and Food Hygiene, College of Public Health, Qingdao University, 308 Ningxia Road, Qingdao 266071, China
| | - Hui Liang
- Department of Nutrition and Food Hygiene, College of Public Health, Qingdao University, 308 Ningxia Road, Qingdao 266071, China
| | - Huaqi Zhang
- Department of Nutrition and Food Hygiene, College of Public Health, Qingdao University, 308 Ningxia Road, Qingdao 266071, China
| |
Collapse
|
23
|
Jing J, Yin S, Liu Y, Liu Y, Wang L, Tang J, Jia G, Liu G, Tian G, Chen X, Cai J, Kang B, Zhao H. Hydroxy Selenomethionine Alleviates Hepatic Lipid Metabolism Disorder of Pigs Induced by Dietary Oxidative Stress via Relieving the Endoplasmic Reticulum Stress. Antioxidants (Basel) 2022; 11:552. [PMID: 35326202 PMCID: PMC8945048 DOI: 10.3390/antiox11030552] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Revised: 03/10/2022] [Accepted: 03/11/2022] [Indexed: 11/18/2022] Open
Abstract
This study used 40 castrated male pigs to determine the protective effects of a new selenium molecule (hydroxy selenomethionine, OH-SeMet) on dietary oxidative stress (DOS) induced hepatic lipid metabolism disorder, and corresponding response of selenotranscriptome. The pigs were randomly grouped into 5 dietary treatments and fed a basal diet formulated with either normal corn and oils or oxidized diet in which the normal corn and oils were replaced by aged corn and oxidized oils, and supplemented with OH-SeMet at 0.0, 0.3, 0.6 and 0.9 mg Se/kg for a period of 16 weeks (n = 8). The results showed that DOS induced liver damage, increased serum alanine aminotransferase (ALT) and alkaline phosphatase (ALP) levels, decreased serum triacylglycerol (TG) level, suppressed antioxidant capacity in the liver, and changed lipid metabolism enzyme activity, thus causing lipid metabolism disorder in the liver. The DOS-induced lipid metabolism disorder was accompanied with endoplasmic reticulum (ER) stress, changes in lipid metabolism-related genes and selenotranscriptome in the liver. Dietary Se supplementation partially alleviated the negative impact of DOS on the lipid metabolism. These improvements were accompanied by increases in Se concentration, liver index, anti-oxidative capacity, selenotranscriptome especially 11 selenoprotein-encoding genes, and protein abundance of GPX1, GPX4 and SelS in the liver, as well as the decrease in SelF abundance. The Se supplementation also alleviated ER stress, restored liver lipid metabolism enzyme activity, increased the mRNA expression of lipid synthesis-related genes, and decreased the mRNA levels of lipidolysis-related genes. In conclusion, the dietary Se supplementation restored antioxidant capacity and mitigated ER stress induced by DOS, thus resisting hepatic lipid metabolism disorders that are associated with regulation of selenotranscriptome.
Collapse
Affiliation(s)
- Jinzhong Jing
- Key Laboratory for Animal Disease-Resistance Nutrition of Ministry of Education, China Ministry of Agriculture and Rural Affairs of Sichuan Province, Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, China; (J.J.); (S.Y.); (Y.L.); (L.W.); (J.T.); (G.J.); (G.L.); (G.T.); (X.C.); (J.C.)
| | - Shenggang Yin
- Key Laboratory for Animal Disease-Resistance Nutrition of Ministry of Education, China Ministry of Agriculture and Rural Affairs of Sichuan Province, Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, China; (J.J.); (S.Y.); (Y.L.); (L.W.); (J.T.); (G.J.); (G.L.); (G.T.); (X.C.); (J.C.)
| | - Yan Liu
- Key Laboratory for Animal Disease-Resistance Nutrition of Ministry of Education, China Ministry of Agriculture and Rural Affairs of Sichuan Province, Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, China; (J.J.); (S.Y.); (Y.L.); (L.W.); (J.T.); (G.J.); (G.L.); (G.T.); (X.C.); (J.C.)
| | - Yonggang Liu
- Adisseo Asia Pacific Pte. Ltd., Singapore 188778, Singapore;
| | - Longqiong Wang
- Key Laboratory for Animal Disease-Resistance Nutrition of Ministry of Education, China Ministry of Agriculture and Rural Affairs of Sichuan Province, Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, China; (J.J.); (S.Y.); (Y.L.); (L.W.); (J.T.); (G.J.); (G.L.); (G.T.); (X.C.); (J.C.)
| | - Jiayong Tang
- Key Laboratory for Animal Disease-Resistance Nutrition of Ministry of Education, China Ministry of Agriculture and Rural Affairs of Sichuan Province, Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, China; (J.J.); (S.Y.); (Y.L.); (L.W.); (J.T.); (G.J.); (G.L.); (G.T.); (X.C.); (J.C.)
| | - Gang Jia
- Key Laboratory for Animal Disease-Resistance Nutrition of Ministry of Education, China Ministry of Agriculture and Rural Affairs of Sichuan Province, Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, China; (J.J.); (S.Y.); (Y.L.); (L.W.); (J.T.); (G.J.); (G.L.); (G.T.); (X.C.); (J.C.)
| | - Guangmang Liu
- Key Laboratory for Animal Disease-Resistance Nutrition of Ministry of Education, China Ministry of Agriculture and Rural Affairs of Sichuan Province, Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, China; (J.J.); (S.Y.); (Y.L.); (L.W.); (J.T.); (G.J.); (G.L.); (G.T.); (X.C.); (J.C.)
| | - Gang Tian
- Key Laboratory for Animal Disease-Resistance Nutrition of Ministry of Education, China Ministry of Agriculture and Rural Affairs of Sichuan Province, Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, China; (J.J.); (S.Y.); (Y.L.); (L.W.); (J.T.); (G.J.); (G.L.); (G.T.); (X.C.); (J.C.)
| | - Xiaoling Chen
- Key Laboratory for Animal Disease-Resistance Nutrition of Ministry of Education, China Ministry of Agriculture and Rural Affairs of Sichuan Province, Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, China; (J.J.); (S.Y.); (Y.L.); (L.W.); (J.T.); (G.J.); (G.L.); (G.T.); (X.C.); (J.C.)
| | - Jingyi Cai
- Key Laboratory for Animal Disease-Resistance Nutrition of Ministry of Education, China Ministry of Agriculture and Rural Affairs of Sichuan Province, Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, China; (J.J.); (S.Y.); (Y.L.); (L.W.); (J.T.); (G.J.); (G.L.); (G.T.); (X.C.); (J.C.)
| | - Bo Kang
- College of Animal Science and Technology, Sichuan Agricultural University, Chengdu 611130, China;
| | - Hua Zhao
- Key Laboratory for Animal Disease-Resistance Nutrition of Ministry of Education, China Ministry of Agriculture and Rural Affairs of Sichuan Province, Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, China; (J.J.); (S.Y.); (Y.L.); (L.W.); (J.T.); (G.J.); (G.L.); (G.T.); (X.C.); (J.C.)
| |
Collapse
|
24
|
Osipova D, Kokoreva K, Lazebnik L, Golovanova E, Pavlov C, Dukhanin A, Orlova S, Starostin K. Regression of Liver Steatosis Following Phosphatidylcholine Administration: A Review of Molecular and Metabolic Pathways Involved. Front Pharmacol 2022; 13:797923. [PMID: 35359878 PMCID: PMC8960636 DOI: 10.3389/fphar.2022.797923] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Accepted: 02/08/2022] [Indexed: 12/21/2022] Open
Abstract
Liver steatosis is a key pathology in non-alcoholic or metabolic associated fatty liver disease. Though largely ignored for decades it is currently becoming the focus of research in hepatology. It is important to consider its origin and current opportunities in terms of pharmacotherapy. Essential phospholipids (EPLs) rich in phosphatidylcholine (PCH) is a widely used treatment option for fatty liver disease, and there is a solid amount of consistent clinical evidence for the regression of steatosis after treatment with EPLs. As knowledge of PCH (a key component of EPLs) pharmacodynamics and mode of action driving this widely observed clinical effect is currently insufficient, we aimed to explore the potential molecular and metabolic pathways involved in the positive effects of PCH on steatosis regression.
Collapse
Affiliation(s)
- D. Osipova
- Research Centre for Medical Genetics, Moscow, Russia
| | - K. Kokoreva
- Institute of Pediatric Endocrinology, Endocrinology Research Centre, Moscow, Russia
| | - L. Lazebnik
- A. I. Evdokimov Moscow State University of Medicine and Dentistry, Ministry of Health of Russia, Moscow, Russia
| | - E. Golovanova
- A. I. Evdokimov Moscow State University of Medicine and Dentistry, Ministry of Health of Russia, Moscow, Russia
| | - Ch. Pavlov
- I. M. Sechenov First Moscow State Medical University, Ministry of Health of Russia, Moscow, Russia
| | - A. Dukhanin
- Molecular Pharmacology and Radiology Department, Russian National Research Medical University, Moscow, Russia
| | - S. Orlova
- Department of Dietetics and Clinical Nutrition of Continuing Medical Education, Medical Institute, RUDN University, Moscow, Russia
| | | |
Collapse
|
25
|
Madduma Hewage S, Au-Yeung KKW, Prashar S, Wijerathne CUB, O K, Siow YL. Lingonberry Improves Hepatic Lipid Metabolism by Targeting Notch1 Signaling. Antioxidants (Basel) 2022; 11:antiox11030472. [PMID: 35326122 PMCID: PMC8944850 DOI: 10.3390/antiox11030472] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Revised: 02/19/2022] [Accepted: 02/25/2022] [Indexed: 02/04/2023] Open
Abstract
Impaired hepatic lipid metabolism is a hallmark of non-alcoholic fatty liver disease (NAFLD), which has no effective treatment option. Recently, Notch signaling has been identified as an important mediator of hepatic lipid metabolism. Lingonberry (Vaccinium vitis-idaea L.) is an anthocyanin-rich fruit with significant lipid-lowering properties. In this study, we examined how lingonberry influenced Notch signaling and fatty acid metabolism in a mouse model of NAFLD. Mice (C57BL/6J) fed a high-fat diet (HFD) for 12 weeks developed fatty liver and activated hepatic Notch1 signaling. Lingonberry supplementation inhibited hepatic Notch1 signaling and improved lipid profile by improving the expression of the genes involved in hepatic lipid metabolism. The results were verified using a palmitic-acid-challenged cell model. Similar to the animal data, palmitic acid impaired cellular lipid metabolism and induced Notch1 in HepG2 cells. Lingonberry extract or cyanidin-3-glucoside attenuated Notch1 signaling and decreased intracellular triglyceride accumulation. The inhibition of Notch in the hepatocytes attenuated sterol-regulatory-element-binding-transcription-factor-1 (SREBP-1c)-mediated lipogenesis and increased the expression of carnitine palmitoyltransferase-I-alpha (CPTIα) and acyl-CoA oxidase1 (ACOX1). Taken together, lingonberry’s hepatoprotective effect is mediated by, in part, improving hepatic lipid metabolism via inhibiting Notch1 signaling in HFD-induced fatty liver.
Collapse
Affiliation(s)
- Susara Madduma Hewage
- Canadian Centre for Agri-Food Research in Health and Medicine, St. Boniface Hospital Research Centre, Winnipeg, MB R2H 2A6, Canada; (S.M.H.); (K.K.W.A.-Y.); (S.P.); (C.U.B.W.)
- Department of Physiology & Pathophysiology, University of Manitoba, Winnipeg, MB R3E 0J9, Canada
| | - Kathy K. W. Au-Yeung
- Canadian Centre for Agri-Food Research in Health and Medicine, St. Boniface Hospital Research Centre, Winnipeg, MB R2H 2A6, Canada; (S.M.H.); (K.K.W.A.-Y.); (S.P.); (C.U.B.W.)
- Department of Animal Science, University of Manitoba, Winnipeg, MB R3T 2N2, Canada
| | - Suvira Prashar
- Canadian Centre for Agri-Food Research in Health and Medicine, St. Boniface Hospital Research Centre, Winnipeg, MB R2H 2A6, Canada; (S.M.H.); (K.K.W.A.-Y.); (S.P.); (C.U.B.W.)
- Agriculture and Agri-Food Canada, St. Boniface Hospital Research Centre, Winnipeg, MB R2H 2A6, Canada
| | - Charith U. B. Wijerathne
- Canadian Centre for Agri-Food Research in Health and Medicine, St. Boniface Hospital Research Centre, Winnipeg, MB R2H 2A6, Canada; (S.M.H.); (K.K.W.A.-Y.); (S.P.); (C.U.B.W.)
- Department of Animal Science, University of Manitoba, Winnipeg, MB R3T 2N2, Canada
| | - Karmin O
- Canadian Centre for Agri-Food Research in Health and Medicine, St. Boniface Hospital Research Centre, Winnipeg, MB R2H 2A6, Canada; (S.M.H.); (K.K.W.A.-Y.); (S.P.); (C.U.B.W.)
- Department of Physiology & Pathophysiology, University of Manitoba, Winnipeg, MB R3E 0J9, Canada
- Department of Animal Science, University of Manitoba, Winnipeg, MB R3T 2N2, Canada
- Correspondence: (K.O.); or (Y.L.S.)
| | - Yaw L. Siow
- Canadian Centre for Agri-Food Research in Health and Medicine, St. Boniface Hospital Research Centre, Winnipeg, MB R2H 2A6, Canada; (S.M.H.); (K.K.W.A.-Y.); (S.P.); (C.U.B.W.)
- Department of Physiology & Pathophysiology, University of Manitoba, Winnipeg, MB R3E 0J9, Canada
- Agriculture and Agri-Food Canada, St. Boniface Hospital Research Centre, Winnipeg, MB R2H 2A6, Canada
- Correspondence: (K.O.); or (Y.L.S.)
| |
Collapse
|
26
|
Dalbeni A, Castelli M, Zoncapè M, Minuz P, Sacerdoti D. Platelets in Non-alcoholic Fatty Liver Disease. Front Pharmacol 2022; 13:842636. [PMID: 35250588 PMCID: PMC8895200 DOI: 10.3389/fphar.2022.842636] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Accepted: 02/03/2022] [Indexed: 12/17/2022] Open
Abstract
Non alcoholic steatohepatitis (NASH) is the inflammatory reaction of the liver to excessive accumulation of lipids in the hepatocytes. NASH can progress to cirrhosis and hepatocellular carcinoma (HCC). Fatty liver is the hepatic manifestation of metabolic syndrome. A subclinical inflammatory state is present in patients with metabolic alterations like insulin resistance, type-2 diabetes, obesity, hyperlipidemia, and hypertension. Platelets participate in immune cells recruitment and cytokines-induced liver damage. It is hypothesized that lipid toxicity cause accumulation of platelets in the liver, platelet adhesion and activation, which primes the immunoinflammatory reaction and activation of stellate cells. Recent data suggest that antiplatelet drugs may interrupt this cascade and prevent/improve NASH. They may also improve some metabolic alterations. The pathophysiology of inflammatory liver disease and the implication of platelets are discussed in details.
Collapse
Affiliation(s)
- Andrea Dalbeni
- Division of General Medicine C, Department of Medicine, University and Azienda Ospedaliera Universitaria Integrata of Verona, Verona, Italy
- Liver Unit, Department of Medicine, University and Azienda Ospedaliera Universitaria Integrata of Verona, Verona, Italy
| | - Marco Castelli
- Division of General Medicine C, Department of Medicine, University and Azienda Ospedaliera Universitaria Integrata of Verona, Verona, Italy
| | - Mirko Zoncapè
- Division of General Medicine C, Department of Medicine, University and Azienda Ospedaliera Universitaria Integrata of Verona, Verona, Italy
- Liver Unit, Department of Medicine, University and Azienda Ospedaliera Universitaria Integrata of Verona, Verona, Italy
| | - Pietro Minuz
- Division of General Medicine C, Department of Medicine, University and Azienda Ospedaliera Universitaria Integrata of Verona, Verona, Italy
- *Correspondence: Pietro Minuz,
| | - David Sacerdoti
- Liver Unit, Department of Medicine, University and Azienda Ospedaliera Universitaria Integrata of Verona, Verona, Italy
| |
Collapse
|
27
|
Xie J, Liu L, Guo H, Bao Q, Hu P, Li H, Che H, Xie W. Orally administered melanin from Sepiapharaonis ink ameliorates depression-anxiety-like behaviors in DSS-induced colitis by mediating inflammation pathway and regulating apoptosis. Int Immunopharmacol 2022; 106:108625. [PMID: 35180627 DOI: 10.1016/j.intimp.2022.108625] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Revised: 01/12/2022] [Accepted: 02/10/2022] [Indexed: 12/20/2022]
Abstract
The effects of intestinal inflammation on the brain and behavior have received a lot of attention. Melanin (MSI) from Sepiapharaonis ink as an emerging functional food, it exhibited a significant protective effect on dextran sulfate sodium (DSS) induced colitis in previous study. In present study, C57BL/6J mice were free to drink 2.5% DSS solution to establish the colitis model. During the DSS treatment, mice were orally administrated with MSI once per day (75, 150, and 300 mg/kg, respectively). The results showed that MSI treatment ameliorated the depression and anxiety symptoms of colitis mice. Further mechanism studies indicated that MSI alleviated inflammatory response by adjusting cytokines TNF-α, IL-1β, IFN-γ, and IL-10, and proteins NLRP3/ASC/caspase-1 inflammasome), inhibited the activation of microglia, restored brain synaptic density, reduced oxidative stress (SOD, MDA) and regulated apoptosis (tunel staining, caspase-3). MSI could modulate depression-anxiety states by targeting inflammation, nerve tissue, oxidative stress and apoptosis. MSI administration could serve as an emerging blue food and nutrition strategy for the prevention of digestive tract inflammation and behavioral disorders.
Collapse
Affiliation(s)
- Jingwen Xie
- College of Marine Science and Biological Engineering, Qingdao University of Science and Technology, Shandong, Qingdao 266042, China
| | - Lin Liu
- College of Marine Science and Biological Engineering, Qingdao University of Science and Technology, Shandong, Qingdao 266042, China
| | - Hao Guo
- College of Marine Science and Biological Engineering, Qingdao University of Science and Technology, Shandong, Qingdao 266042, China
| | - Qi Bao
- College of Marine Science and Biological Engineering, Qingdao University of Science and Technology, Shandong, Qingdao 266042, China
| | - Penglong Hu
- College of Marine Science and Biological Engineering, Qingdao University of Science and Technology, Shandong, Qingdao 266042, China
| | - Hongyan Li
- College of Marine Science and Biological Engineering, Qingdao University of Science and Technology, Shandong, Qingdao 266042, China; Shandong Provincial Key Laboratory of Biochemical Engineering, Shandong, Qingdao 266042, China; Shandong Provincial Key Laboratory of Biochemical Engineering, Shandong, Qingdao 266042, China
| | - Hongxia Che
- College of Marine Science and Biological Engineering, Qingdao University of Science and Technology, Shandong, Qingdao 266042, China; Shandong Provincial Key Laboratory of Biochemical Engineering, Shandong, Qingdao 266042, China; Shandong Provincial Key Laboratory of Biochemical Engineering, Shandong, Qingdao 266042, China
| | - Wancui Xie
- College of Marine Science and Biological Engineering, Qingdao University of Science and Technology, Shandong, Qingdao 266042, China; Shandong Provincial Key Laboratory of Biochemical Engineering, Shandong, Qingdao 266042, China; Shandong Provincial Key Laboratory of Biochemical Engineering, Shandong, Qingdao 266042, China.
| |
Collapse
|
28
|
Fu Y, Gao J, Li Y, Yang X, Zhang Y. RETRACTED: TRIM21 deficiency confers protection from OGD/R-induced oxidative and inflammatory damage in cultured hippocampal neurons through regulation of the Keap1/Nrf2 pathway. Int Immunopharmacol 2022; 103:108414. [PMID: 34929478 DOI: 10.1016/j.intimp.2021.108414] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Revised: 11/10/2021] [Accepted: 11/25/2021] [Indexed: 02/06/2023]
Abstract
This article has been retracted: please see Elsevier Policy on Article Withdrawal (http://www.elsevier.com/locate/withdrawalpolicy). The authors have requested that this paper be retracted as they were unable to repeat some results reported in this paper under the same conditions. In Figure 1D, they found that TRIM21 siRNA-1 could not silence the expression of TIRM21. Therefore, the subsequent results were no longer reliable. The authors apologize for any inconvenience this retraction may cause for readers.
Collapse
Affiliation(s)
- Yahong Fu
- Department of Neurology, Xi'an Ninth Hospital, No. 151 East Section of South Second Ring Road, Xi'an 710054, Shaanxi Province, China
| | - Junxian Gao
- Department of Neurology, Xi'an Ninth Hospital, No. 151 East Section of South Second Ring Road, Xi'an 710054, Shaanxi Province, China
| | - Yanqing Li
- Department of Neurology, Xi'an Ninth Hospital, No. 151 East Section of South Second Ring Road, Xi'an 710054, Shaanxi Province, China
| | - Xi Yang
- Department of Neurology, Xi'an Ninth Hospital, No. 151 East Section of South Second Ring Road, Xi'an 710054, Shaanxi Province, China
| | - Yun Zhang
- Department of Neurology, Xi'an Ninth Hospital, No. 151 East Section of South Second Ring Road, Xi'an 710054, Shaanxi Province, China.
| |
Collapse
|
29
|
Probiotic Bacillus Alleviates Oxidative Stress-Induced Liver Injury by Modulating Gut-Liver Axis in a Rat Model. Antioxidants (Basel) 2022; 11:antiox11020291. [PMID: 35204173 PMCID: PMC8868294 DOI: 10.3390/antiox11020291] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2022] [Revised: 01/27/2022] [Accepted: 01/27/2022] [Indexed: 12/12/2022] Open
Abstract
Emerging evidence suggests a key role of gut microbiota in maintaining liver functions through modulating the gut–liver axis. In this study, we investigated whether microbiota alteration mediated by probiotic Bacillus was involved in alleviating oxidative stress- induced liver injury. Sprague–Dawley rats were orally administered Bacillus SC06 or SC08 for a 24-day period and thereafter intraperitoneally injected diquat (DQ) to induce oxidative stress. Results showed that Bacillus, particularly SC06 significantly inhibited hepatic injuries, as evidenced by the alleviated damaged liver structure, the decreased levels of ALT, AST, ALP and LDH, and the suppressed mitochondrial dysfunction. SC06 pretreatment markedly enhanced the liver antioxidant capacity by decreasing MDA and p47, and increasing T-AOC, SOD and HO-1.16S rRNA sequencing analysis revealed that DQ significantly changed the diversities and composition of gut microbiota, whereas Bacillus pretreatments could attenuate gut dysbiosis. Pearson’s correlation analysis showed that AST and MDA exerted a positive correlation with the opportunistic pathogenic genera and species (Escherichia and Shigella), and negatively correlated with the potential probiotics (Lactobacillus), while SOD exerted a reverse trend. The microbial metagenomic analysis demonstrated that Bacillus, particularly SC06 markedly suppress the metabolic pathways such as carbohydrate metabolism, lipid metabolism, amino acid metabolism and metabolism of cofactors and vitamins. Furthermore, SC06 decreased the gene abundance of the pathways mediating bacterial replication, secretion and pathogenicity. Taken together, Bacillus SC06 alleviates oxidative stress-induced liver injuries via optimizing the composition, metabolic pathways and pathogenic replication and secretion of gut microbiota. These findings elucidate the mechanisms of probiotics in alleviating oxidative stress and provide a promising strategy for preventing liver diseases by targeting gut microbiota.
Collapse
|
30
|
Pukhalskaia AE, Diatlova AS, Linkova NS, Kvetnoy IM. Sirtuins: Role in the Regulation of Oxidative Stress and the Pathogenesis of Neurodegenerative Diseases. NEUROSCIENCE AND BEHAVIORAL PHYSIOLOGY 2022; 52:164-174. [DOI: 10.1007/s11055-022-01217-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Revised: 08/23/2020] [Accepted: 09/28/2020] [Indexed: 01/03/2025]
|
31
|
Zhu F, Shao J, Tian Y, Xu Z. Sulfiredoxin-1 protects retinal ganglion cells from high glucose-induced oxidative stress and inflammatory injury by potentiating Nrf2 signaling via the Akt/GSK-3β pathway. Int Immunopharmacol 2021; 101:108221. [PMID: 34653733 DOI: 10.1016/j.intimp.2021.108221] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Revised: 09/17/2021] [Accepted: 09/30/2021] [Indexed: 12/12/2022]
Abstract
Sulfiredoxin-1 (Srxn1) has been acknowledged as a remarkable pro-survival factor in the protection of cells against stress-induced damage. The persistent exposure of retinal ganglion cells (RGCs) to high glucose (HG) in diabetes induces cellular damage, which contributes to the onset of diabetic retinopathy, a severe complication of diabetes. So far, little is known about the role of Srxn1 in regulating HG-induced injury of RGCs. The goals of this work were to evaluate the possible relevance of Srxn1 in the modulation of HG-induced apoptosis, oxidative stress and inflammation of RGCs in vitro. Our data showed that HG exposure caused a marked decrease in Srxn1 expression in RGCs. The up-regulation of Srxn1 markedly decreased HG-evoked apoptosis, reactive oxygen species (ROS) generation and pro-inflammatory cytokine release in RGCs. On the contrary, the depletion of Srxn1 rendered RGCs more susceptible to HG-induced injury. Further data demonstrated that Srnx1 enhanced the activation of nuclear factor erythroid-2 (E2)-related factor 2 (Nrf2) signaling in HG-exposed RGCs associated with up-regulating the phosphorylation of Akt and glucogen synthase kinase-3β (GSK-3β). Notably, the inhibition of Akt abolished Srnx1-overexpression-mediated Nrf2 activation, while GSK-3β inhibition reversed Srnx1-depletion-mediated inactivation of Nrf2. In addition, Nrf2 inhibition partially abrogated Srnx1-mediated protective effects against HG-induced injury of RGCs. In summary, these data demonstrate that the overexpression of Srxn1 protects RGCs from the HG-induced injury of RGCs by enhancing Nrf2 signaling via modulation of Akt/GSK-3β axis. Our work highlights that the Srxn1-mediated Akt/GSK-3β/Nrf2 axis may exert a possible role in regulating RGC injury of diabetic retinopathy.
Collapse
Affiliation(s)
- Fei Zhu
- Ophthalmology, Yulin Hospital of Traditional Chinese Medicine, Yulin 719000, China
| | - Juan Shao
- Ophthalmology, Shaanxi Eye Hospital, Xi'an People's Hospital (Xi'an Fourth Hospital), Affiliated Guangren Hospital, School of Medicine, Xi'an Jiaotong University, Xi'an 710004, China.
| | - Yunlin Tian
- Ophthalmology, Shaanxi Eye Hospital, Xi'an People's Hospital (Xi'an Fourth Hospital), Affiliated Guangren Hospital, School of Medicine, Xi'an Jiaotong University, Xi'an 710004, China
| | - Zhiguo Xu
- Ophthalmology, Shaanxi Eye Hospital, Xi'an People's Hospital (Xi'an Fourth Hospital), Affiliated Guangren Hospital, School of Medicine, Xi'an Jiaotong University, Xi'an 710004, China
| |
Collapse
|
32
|
Holzner LMW, Murray AJ. Hypoxia-Inducible Factors as Key Players in the Pathogenesis of Non-alcoholic Fatty Liver Disease and Non-alcoholic Steatohepatitis. Front Med (Lausanne) 2021; 8:753268. [PMID: 34692739 PMCID: PMC8526542 DOI: 10.3389/fmed.2021.753268] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Accepted: 09/10/2021] [Indexed: 12/20/2022] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) and its more severe form non-alcoholic steatohepatitis (NASH) are a major public health concern with high and increasing global prevalence, and a significant disease burden owing to its progression to more severe forms of liver disease and the associated risk of cardiovascular disease. Treatment options, however, remain scarce, and a better understanding of the pathological and physiological processes involved could enable the development of new therapeutic strategies. One process implicated in the pathology of NAFLD and NASH is cellular oxygen sensing, coordinated largely by the hypoxia-inducible factor (HIF) family of transcription factors. Activation of HIFs has been demonstrated in patients and mouse models of NAFLD and NASH and studies of activation and inhibition of HIFs using pharmacological and genetic tools point toward important roles for these transcription factors in modulating central aspects of the disease. HIFs appear to act in several cell types in the liver to worsen steatosis, inflammation, and fibrosis, but may nevertheless improve insulin sensitivity. Moreover, in liver and other tissues, HIF activation alters mitochondrial respiratory function and metabolism, having an impact on energetic and redox homeostasis. This article aims to provide an overview of current understanding of the roles of HIFs in NAFLD, highlighting areas where further research is needed.
Collapse
Affiliation(s)
| | - Andrew J. Murray
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, United Kingdom
| |
Collapse
|
33
|
Sirtuins and Renal Oxidative Stress. Antioxidants (Basel) 2021; 10:antiox10081198. [PMID: 34439446 PMCID: PMC8388938 DOI: 10.3390/antiox10081198] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 07/04/2021] [Accepted: 07/23/2021] [Indexed: 02/06/2023] Open
Abstract
Renal failure is a major health problem that is increasing worldwide. To improve clinical outcomes, we need to understand the basic mechanisms of kidney disease. Aging is a risk factor for the development and progression of kidney disease. Cells develop an imbalance of oxidants and antioxidants as they age, resulting in oxidative stress and the development of kidney damage. Calorie restriction (CR) is recognized as a dietary approach that promotes longevity, reduces oxidative stress, and delays the onset of age-related diseases. Sirtuins, a type of nicotinamide adenine dinucleotide (NAD)-dependent histone deacetylase, are considered to be anti-aging molecules, and CR induces their expression. The sirtuin family consists of seven enzymes (Sirt1–7) that are involved in processes and functions related to antioxidant and oxidative stress, such as DNA damage repair and metabolism through histone and protein deacetylation. In fact, a role for sirtuins in the regulation of antioxidants and redox substances has been suggested. Therefore, the activation of sirtuins in the kidney may represent a novel therapeutic strategy to enhancing resistance to many causative factors in kidney disease through the reduction of oxidative stress. In this review, we discuss the relationship between sirtuins and oxidative stress in renal disease.
Collapse
|
34
|
Luo H, Xu N, Wu J, Gan Y, Chen L, Guan F, Li M, Li Y, Chen J, Su Z, Liu Y. β-patchoulene protects against non-alcoholic steatohepatitis via interrupting the vicious circle among oxidative stress, histanoxia and lipid accumulation in rats. Int Immunopharmacol 2021; 98:107915. [PMID: 34198236 DOI: 10.1016/j.intimp.2021.107915] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Revised: 06/18/2021] [Accepted: 06/21/2021] [Indexed: 01/06/2023]
Abstract
Non-alcoholic steatohepatitis (NASH), an extreme progressive subtype of metabolic associated fatty liver disease, is well characterized by hepatic steatosis, injury and inflammation. It causes irreversible hepatic damage and there are no approved interventions for it. β-PAE, a representatively pharmacological active substance isolated from Pogostemon cablin, has been indicated to alleviate hepatic steatosis and injury through modulating lipid metabolism in rats with simple steatosis. However, its protection against NASH remains unclear. Here, this study explored the potential effect of β-PAE against high-fat diet-induced NASH in rats. The results displayed that β-PAE significantly reduced the gains of body weight and epididymal adipose tissue, liver index and attenuated liver histological damages in NASH rats. It also markedly alleviated hepatic inflammation by inhibiting NLRP3 inflammasome activation. In NASH, the active NLRP3 inflammasome is caused by hepatic lipid abnormal accumulation-induced oxidative stress. Excessive oxidative stress results in hepatic histanoxia, which exacerbates lipid metabolism disorders by elevating CD36 to suppress AMPK signalling pathways. Moreover, the lipid accumulation led by lipid metabolism dysfunction intensifies oxidative stress. A vicious circle is formed among oxidative stress, histanoxia and lipid accumulation, eventually, but β-PAE effectively interrupted it. Interestingly, soluble CD36 (sCD36) was tightly associated not only with hepatic steatosis and injury but also with inflammation. Collectively, β-PAE exerted a positive effect against NASH by interrupting the vicious circle among oxidative stress, histanoxia and lipid accumulation, and sCD36 may be a promising non-invasive tool for NASH diagnosis.
Collapse
Affiliation(s)
- Huijuan Luo
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou 510006, China
| | - Nan Xu
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou 510006, China
| | - Jiazhen Wu
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou 510006, China
| | - Yuxuan Gan
- The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou 510120, China
| | - Liping Chen
- Faculty of Health Sciences, University of Macau, Macao, China
| | - Fengkun Guan
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou 510006, China
| | - Mengyao Li
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou 510006, China
| | - Yucui Li
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou 510006, China
| | - Jiannan Chen
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou 510006, China
| | - Ziren Su
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou 510006, China; Dongguan & Guangzhou University of Chinese Medicine Cooperative Academy of Mathematical Engineering for Chinese Medicine, Dongguan 523808, China.
| | - Yuhong Liu
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou 510006, China; Dongguan & Guangzhou University of Chinese Medicine Cooperative Academy of Mathematical Engineering for Chinese Medicine, Dongguan 523808, China.
| |
Collapse
|
35
|
Dey R, Bishayi B. Ciprofloxacin and dexamethasone in combination attenuate S. aureus induced brain abscess via neuroendocrine-immune interaction of TLR-2 and glucocorticoid receptor leading to behavioral improvement. Int Immunopharmacol 2021; 97:107695. [PMID: 33962227 DOI: 10.1016/j.intimp.2021.107695] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Revised: 04/06/2021] [Accepted: 04/18/2021] [Indexed: 12/14/2022]
Abstract
Staphylococcus aureus induced brain abscess is a critical health concern throughout the developing world. The conventional surgical intervention could not regulate the abscess-induced brain inflammation. Thus further study over the alternative therapeutic strategy for treating a brain abscess is of high priority. The resident glial cells recognize the invading S. aureus by their cell surface Toll-like receptor-2 (TLR-2). Glucocorticoid receptor (GR) was known for its immunosuppressive effects. In this study, an attempt had been taken to utilize the functional relationship or cross-talking between TLR-2 and GR during the pathogenesis of brain abscesses. Here, the combination of an antibiotic (i.e. ciprofloxacin) and dexamethasone was used to regulate the brain inflammation either in TLR-2 or GR blocking condition. We were also interested to figure out the possible impact of alternative therapy on behavioral impairments. The results indicated that combination treatment during TLR-2 blockade significantly reduced the bacterial burden and abscess area score in the infected brain. However, marked improvements were observed in anxiety, depression-like behavior, and motor co-ordination. The combination treatment after TLR-2 blocking effectively scavenged free radicals (H2O2, superoxide anion, and NO) through modulating antioxidant enzyme activities that ultimately control S. aureus induced glial reactivity possibly via up-regulating GR expression. The exogenous dexamethasone might regulate the GR expression in the brain by increasing the corticosterone concentration and the GC-GR mediated signaling. Therefore, this in-vivo study demonstrates the possible regulatory mechanism of bacterial brain abscess that involved TLR-2 and GR as a part of neuroendocrine-immune interaction.
Collapse
Affiliation(s)
- Rajen Dey
- Department of Physiology, Immunology Laboratory, University of Calcutta, University Colleges of Science and Technology, Calcutta, West Bengal, India
| | - Biswadev Bishayi
- Department of Physiology, Immunology Laboratory, University of Calcutta, University Colleges of Science and Technology, Calcutta, West Bengal, India.
| |
Collapse
|
36
|
Oğul Y, Gür F, Cengiz M, Gür B, Sarı RA, Kızıltunç A. Evaluation of oxidant and intracellular anti-oxidant activity in rheumatoid arthritis patients: In vivo and in silico studies. Int Immunopharmacol 2021; 97:107654. [PMID: 33895477 DOI: 10.1016/j.intimp.2021.107654] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Revised: 03/20/2021] [Accepted: 04/03/2021] [Indexed: 11/24/2022]
Abstract
Rheumatoid Arthritis (RA) is the most prevalent cause of the systematic inflammatory arthritis that destroys the joints. While the pathogenesis of RA remains to be clarified, the imbalance in the oxidant and anti-oxidant defense system plays a crucial role. This study aims to evaluate oxidant and anti-oxidant levels of RA patients and their impacts on the activity of the disease via in silico studies. 28 patients who had not previously received any treatment for RA and 20 healthy controls were included. Their oxidative stress markers, antioxidant markers, and inflammatory factors were investigated via in silico studies. Compared to the Control Group, serum CRP levels, MDA levels, and XO activities were higher in RA Group. Cu/ZnSOD and GPx activities decreased while CAT activities remained unchanged. Besides, there was a positive correlation between MDA-serum CRP levels but a negative correlation between MDA levels-Cu/ZnSOD activities. Furthermore, we observed a negative correlation between CRP levels and Cu/ZnSOD activities. Based on these results, it was concluded that oxidative stress had increased, the defense system had weakened, and ROS production had increased. Finally, our study results with SOD and CAT activity were confirmed by molecular docking studies.
Collapse
Affiliation(s)
- Yasemin Oğul
- Department of Medicinal Biochemistry, Regional Training and Research Hospital, Health Sciences University, 25240 Erzurum, Turkey.
| | - Fatma Gür
- Department of Medical Services and Techniques, Health Services Vocational School, Ataturk University, 25240 Erzurum, Turkey
| | - Mustafa Cengiz
- Department of Elementary Education, Faculty of Education, Siirt University, 56100 Siirt, Turkey
| | - Bahri Gür
- Department of Biochemistry, Faculty of Sciences and Arts, Iğdır University, 76000 Iğdır, Turkey
| | - Refik Ali Sarı
- Department of Internal Diseases, Faculty of Medicine, Karadeniz Technical University, 61080 Trabzon, Turkey
| | - Ahmet Kızıltunç
- Department of Biochemistry, Faculty of Medicine, Ataturk University, 25240 Erzurum, Turkey
| |
Collapse
|
37
|
Heydari H, Ghiasi R, Ghaderpour S, Keyhanmanesh R. The Mechanisms Involved in Obesity-Induced Male Infertility. Curr Diabetes Rev 2021; 17:259-267. [PMID: 32814535 DOI: 10.2174/1573399816666200819114032] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/17/2020] [Revised: 07/20/2020] [Accepted: 07/29/2020] [Indexed: 11/22/2022]
Abstract
BACKGROUND Obesity resulted by imbalance between the intake of energy and energy consumption can lead to growth and metabolic disease development in people. Both in obese men and animal models, several studies indicate that obesity leads to male infertility. OBJECTIVE This review has discussed some mechanisms involved in obesity-induced male infertility. METHODS Online documents were searched through Science Direct, Pubmed, Scopus, and Google Scholar websites dating from 1959 to recognize studies on obesity, kisspeptin, leptin, and infertility. RESULTS Obesity induced elevated inflammatory cytokines and oxidative stress can affect male reproductive functions, including spermatogenesis disorders, reduced male fertility power and hormones involved in the hypothalamus-pituitary-gonadal axis. CONCLUSION There is significant evidence that obesity resulted in male infertility. Obesity has a negative effect on male reproductive function via several mechanisms such as inflammation and oxidative stress.
Collapse
Affiliation(s)
- Hamed Heydari
- Department of Physiology, Tabriz Faculty of Medical Science, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Rafighe Ghiasi
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Saber Ghaderpour
- Department of Physiology, Tabriz Faculty of Medical Science, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Rana Keyhanmanesh
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
38
|
Camargo TF, Zanesco AM, Pacher KAS, Andrade TAM, Alves AA, do Amaral MEC. Physiological profile regulation during weight gain and loss by ovariectomized females: importance of SIRT1 and SIRT4. Am J Physiol Endocrinol Metab 2020; 319:E769-E778. [PMID: 32865007 DOI: 10.1152/ajpendo.00465.2019] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Obesity in menopausal women occurs because of the systemic effects of loss of ovarian function, resulting in increased body weight and oxidative stress. Caloric restriction (CR) is essential for weight loss, since it provides benefits associated with metabolic normalization resulting from the action of sirtuins. The aim of this work was to evaluate the physiological effects of weight cycling in ovariectomized females. Females aged 2 mo (n = 8/group) were submitted to simulated surgery, ovariectomy (OVX group), and ovariectomy with weight fluctuation (WF group). In the WF group, weight cycling was performed two times, using 21 days of ad libitum commercial feed and 21 days of caloric restriction with 40% of the feed consumed by the OVX group. After 17 wk, the animals were evaluated experimentally. Weight fluctuations reduced triacylglycerol and the adipose tissue index of the WF animals, while increasing the expression of antioxidant proteins. In addition to causing fluctuations in the physiological parameters, the weight cycling led to increases of adipocyte number and serum fatty acids. These effects were reflected in increased expression of the sirtuin (SIRT) 1 and SIRT4 proteins, as well as protein complexes of the mitochondrial electron transport chain, especially in the liver and adipose tissues. The weight-cycling results suggested that mitochondrial and nuclear sirtuins were active in cellular signaling for the control of lipid metabolism, oxidative phosphorylation, and redox status. Weight cycling was able to restore the health characteristics of lean animals.
Collapse
Affiliation(s)
- Thaís Furtado Camargo
- Graduate Program in Biomedical Sciences, Centro Universitário Hermínio Ometto, FHO/UNIARARAS, Araras, Sãu Paulo, Brazil
| | - Ariane Maria Zanesco
- College of Biomedicine, Centro Universitário Hermínio Ometto, Araras, Sãu Paulo, Brazil
| | - Kayo Augusto Salandin Pacher
- Graduate Program in Biomedical Sciences, Centro Universitário Hermínio Ometto, FHO/UNIARARAS, Araras, Sãu Paulo, Brazil
| | | | | | | |
Collapse
|
39
|
Impact of Nutrition on Short-Term Exercise-Induced Sirtuin Regulation: Vegans Differ from Omnivores and Lacto-Ovo Vegetarians. Nutrients 2020; 12:nu12041004. [PMID: 32260570 PMCID: PMC7230533 DOI: 10.3390/nu12041004] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2020] [Revised: 04/01/2020] [Accepted: 04/02/2020] [Indexed: 12/14/2022] Open
Abstract
Keywords: sirtuins; vegetarian; vegan; exercise; endurance athletes; metabolic regulation.
Collapse
|
40
|
Pratt R, Lakhani HV, Zehra M, Desauguste R, Pillai SS, Sodhi K. Mechanistic Insight of Na/K-ATPase Signaling and HO-1 into Models of Obesity and Nonalcoholic Steatohepatitis. Int J Mol Sci 2019; 21:ijms21010087. [PMID: 31877680 PMCID: PMC6982200 DOI: 10.3390/ijms21010087] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Revised: 12/18/2019] [Accepted: 12/20/2019] [Indexed: 02/07/2023] Open
Abstract
Obesity is a multifaceted pathophysiological condition that has been associated with lipid accumulation, adipocyte dysfunction, impaired mitochondrial biogenesis and an altered metabolic profile. Redox imbalance and excessive release of inflammatory mediators have been intricately linked in obesity-associated phenotypes. Hence, understanding the mechanisms of redox signaling pathways and molecular targets exacerbating oxidative stress is crucial in improving health outcomes. The activation of Na/K-ATPase/Src signaling, and its downstream pathways, by reactive oxygen species (ROS) has been recently implicated in obesity and subsequent nonalcoholic steatohepatitis (NASH), which causes further production of ROS creating an oxidant amplification loop. Apart from that, numerous studies have also characterized antioxidant properties of heme oxygenase 1 (HO-1), which is suppressed in an obese state. The induction of HO-1 restores cellular redox processes, which contributes to inhibition of the toxic milieu. The novelty of these independent mechanisms presents a unique opportunity to unravel their potential as molecular targets for redox regulation in obesity and NASH. The attenuation of oxidative stress, by understanding the underlying molecular mechanisms and associated mediators, with a targeted treatment modality may provide for improved therapeutic options to combat clinical disorders.
Collapse
Affiliation(s)
| | | | | | | | | | - Komal Sodhi
- Correspondence: ; Tel.: +1-(304)-691-1704; Fax: +1-(914)-347-4956
| |
Collapse
|
41
|
Li Z, Li H, Zhao ZB, Zhu W, Feng PP, Zhu XW, Gong JP. SIRT4 silencing in tumor-associated macrophages promotes HCC development via PPARδ signalling-mediated alternative activation of macrophages. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2019; 38:469. [PMID: 31744516 PMCID: PMC6862746 DOI: 10.1186/s13046-019-1456-9] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/17/2019] [Accepted: 10/17/2019] [Indexed: 12/12/2022]
Abstract
Background The activation of tumour-associated macrophages (TAMs) contributes to the progression of hepatocellular carcinoma (HCC). SIRT4 acts as a tumour suppressor of tumour growth by regulating cell metabolism, inflammation, and anti-tumourigenesis. However, the involvement of SIRT4 in the activation of TAMs is unknown. Based on previous findings, the expression of SIRT4 in distinct groups of TAMs as well as the effect of SIRT4 silencing on macrophage polarization was investigated. Methods The expression of SIRT4 in HCC tissues and peritumour tissues was tested by qRT-PCR, western blotting and histological analysis. A Kaplan-Meier survival curve was generated based on the expression of SIRT4 in the HCC samples. Next, immunofluorescence staining was used to evaluate distinct groups of TAMs in human HCC samples, and the expression of SIRT4 in M1 and M2 TAMs was examined by flow cytometry. A homograft mouse model was used to assess the effect of SIRT4 silencing in TAMs on the development of HCC cells. Results SIRT4 was significantly downregulated in HCC tumour tissues, and the expression of SIRT4 in peritumour tissues was positively associated with survival in patients. We further found that downregulation of SIRT4 was associated with increased macrophage infiltration and a high ratio of M2/M1 macrophages in HCC peritumour tissues. Using gene interference, we found that SIRT4 silencing in TAMs significantly modulated the alternative activation of macrophages and promoted in vitro and in vivo HCC cell growth. Mechanistically, we revealed that HCM restricted the expression of SIRT4 in macrophages and promoted alternative activation of macrophages via the FAO-PPARδ-STAT3 axis. Furthermore, we also revealed that elevated MCP-1 expression induced by SIRT4 downregulation was responsible for increased TAM infiltration in peritumour tissues. Conclusions Overall, our results demonstrate that downregulation of SIRT4 in TAMs modulates the alternative activation of macrophages and promotes HCC development via the FAO-PPARδ-STAT3 axis. These results could provide a new therapeutic target for the treatment of HCC.
Collapse
Affiliation(s)
- Zhi Li
- Department of Hepatobiliary Surgery, the Second Affiliated Hospital of Chongqing Medical University, Yuzhong District, Chongqing, 400010, People's Republic of China
| | - He Li
- Department of General Surgery, the YongChuan Hospital of Chongqing Medical University, YongChuan District, Chongqing, 402160, People's Republic of China
| | - Zhi-Bo Zhao
- Department of Hepatobiliary Surgery, the Second Affiliated Hospital of Chongqing Medical University, Yuzhong District, Chongqing, 400010, People's Republic of China
| | - Wei Zhu
- Department of Hepatobiliary Surgery, the Second Affiliated Hospital of Chongqing Medical University, Yuzhong District, Chongqing, 400010, People's Republic of China
| | - Pan-Pan Feng
- Department of Hepatobiliary Surgery, the Second Affiliated Hospital of Chongqing Medical University, Yuzhong District, Chongqing, 400010, People's Republic of China
| | - Xi-Wen Zhu
- Department of Hepatobiliary Surgery, the Second Affiliated Hospital of Chongqing Medical University, Yuzhong District, Chongqing, 400010, People's Republic of China
| | - Jian-Ping Gong
- Department of Hepatobiliary Surgery, the Second Affiliated Hospital of Chongqing Medical University, Yuzhong District, Chongqing, 400010, People's Republic of China.
| |
Collapse
|
42
|
Dichotomous Sirtuins: Implications for Drug Discovery in Neurodegenerative and Cardiometabolic Diseases. Trends Pharmacol Sci 2019; 40:1021-1039. [PMID: 31704173 DOI: 10.1016/j.tips.2019.09.003] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2019] [Revised: 09/12/2019] [Accepted: 09/12/2019] [Indexed: 02/06/2023]
Abstract
Sirtuins (SIRT1-7), a class of NAD+-dependent deacylases, are central regulators of metabolic homeostasis and stress responses. While numerous salutary effects associated with sirtuin activation, especially SIRT1, are well documented, other reports show health benefits resulting from sirtuin inhibition. Furthermore, conflicting findings have been obtained regarding the pathophysiological role of specific sirtuin isoforms, suggesting that sirtuins act as 'double-edged swords'. Here, we provide an integrated overview of the different findings on the role of mammalian sirtuins in neurodegenerative and cardiometabolic disorders and attempt to dissect the reasons behind these different effects. Finally, we discuss how addressing these obstacles may provide a better understanding of the complex sirtuin biology and improve the likelihood of identifying effective and selective drug targets for a variety of human disorders.
Collapse
|
43
|
Han Y, Zhou S, Coetzee S, Chen A. SIRT4 and Its Roles in Energy and Redox Metabolism in Health, Disease and During Exercise. Front Physiol 2019; 10:1006. [PMID: 31447696 PMCID: PMC6695564 DOI: 10.3389/fphys.2019.01006] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2019] [Accepted: 07/22/2019] [Indexed: 01/25/2023] Open
Abstract
NAD+-dependent SIRT4 has been reported to be a key regulator of metabolic enzymes and antioxidant defense mechanisms in mitochondria. It also plays an important role in regulation of mitochondrial metabolism in response to exercise. Recent studies have shown that SIRT4 is involved in a wide range of mitochondrial metabolic processes, including depressing insulin secretion in pancreatic beta cells, promoting lipid synthesis, regulating mitochondrial adenosine triphosphate (ATP) homeostasis, controlling apoptosis and regulating redox. SIRT4 also appears to have enzymatic functions involved in posttranslational modifications such as ADP-ribosylation, lysine deacetylation and lipoamidation. However, the effects on SIRT4 by metabolic diseases and changes in metabolic homeostasis such as during exercise, along with the roles of SIRT4 in the regulation of metabolism during disease, are not well understood. The main goal of this review is to critically analyse and summarise the current research evidence on the significance of the SIRT4 as a metabolic regulator and in mitochondrial function and its putative roles in relation to metabolic diseases and exercise.
Collapse
Affiliation(s)
- Yumei Han
- School of Physical Education, Shanxi University, Taiyuan, China
| | - Shi Zhou
- School of Health and Human Sciences, Southern Cross University, Lismore, NSW, Australia
| | - Sonja Coetzee
- School of Health and Human Sciences, Southern Cross University, Lismore, NSW, Australia
| | - Anping Chen
- School of Physical Education, Shanxi University, Taiyuan, China
| |
Collapse
|
44
|
Ji L, Chen Y, Wang H, Zhang W, He L, Wu J, Liu Y. Overexpression of Sirt6 promotes M2 macrophage transformation, alleviating renal injury in diabetic nephropathy. Int J Oncol 2019; 55:103-115. [PMID: 31115579 PMCID: PMC6561622 DOI: 10.3892/ijo.2019.4800] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2018] [Accepted: 03/01/2019] [Indexed: 02/06/2023] Open
Abstract
In this study, we aimed to investigate the associations between Sirt6, macrophages and diabetic nephropathy (DN). Immunohistochemical, western blot and RT-qPCR analyses were performed to detect the expression levels of Sirt6, the markers of podocytes and monocytes and related inflammatory factors in the tissues of rats with streptozocin-induced DN. A series of cell experiments in isolated culture or the co-culture of macrophages and podocytes were conducted to examine the effects of the overexpression of Sirt6 on macrophage transformation, podocyte apoptosis and associated genes, and analyses were performed using RT-qPCR, flow cytometry and western blot analysis, where appropriate. In the rat model of DN, injured podocytes were represented by the decreased protein expression levels of Nephrin and Sirt6, and by an increased Desmin expression. Additionally, the M1 phenotype transformation of macrophages was evidenced by the increased expression levels of CD86, tumor necrosis factor (TNF)-α and inducible nitric oxide synthase (iNOS), and by the decreased expression levels of CD206, Sirt6, interleukin (IL)-4 and IL-10. In vitro assays of macrophages and podocytes demonstrated that glucose promoted macrophage M1 transformation and podocyte apoptosis in a dose-dependent manner and attenuated Sirt6 expression. Macrophages transformed into the M2 phenotype following the overexpression of Sirt6 by the successful transfection of macrophages with a Sirt6 overexpression plasmid. Sirt6 was also overexpressed in podocytes. In a Transwell co-culture system, the overexpression of Sirt6 in macrophages (but not the overexpression of Sirt6 in podocytes) protected the podocytes from high-glucose-induced injury. However, the apoptosis of the podocytes overexpressing Sirt6 (induced by transfection with a Sirt6 overexpression plasmid) still increased when these podocytes were co-cultured with macrophages in high-glucose medium. These protective effects were evidenced by the inhibition of apoptosis, the upregulation of the expression levels of Bcl-2 and CD206, as well as by the decreased expression levels of Bax and CD86. On the whole, the findings of this study suggest that Sirt6 protects podocytes against injury in a mimicked diabetic kidney microenvironment by activating M2 macrophages, indicating that Sirt6 can act as an immune response regulatory factor in DN-associated renal inflammatory injury.
Collapse
Affiliation(s)
- Liqiang Ji
- Department of Pharmacy, The First People's Hospital of Yuhang District, Hangzhou, Zhejiang 311100, P.R China
| | - Yifang Chen
- Department of Pharmacy, The First People's Hospital of Yuhang District, Hangzhou, Zhejiang 311100, P.R China
| | - Hongqiang Wang
- Department of Pharmacy, Hangzhou Yuhang TCM Hospital, Hangzhou, Zhejiang 311106, P.R China
| | - Wei Zhang
- Department of Pharmacy, The First People's Hospital of Yuhang District, Hangzhou, Zhejiang 311100, P.R China
| | - Lanxiang He
- Department of Pharmacy, The First People's Hospital of Yuhang District, Hangzhou, Zhejiang 311100, P.R China
| | - Jingmin Wu
- Department of Pharmacy, The First People's Hospital of Yuhang District, Hangzhou, Zhejiang 311100, P.R China
| | - Yinghui Liu
- Department of Endocrinology, Zhejiang Provincial Integrated Chinese and Western Medicine Hospital, Hangzhou Red Cross Hospital, Hangzhou, Zhejiang 310003, P.R China
| |
Collapse
|
45
|
Higd1a Protects Cells from Lipotoxicity under High-Fat Exposure. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2019; 2019:6051262. [PMID: 31089410 PMCID: PMC6476072 DOI: 10.1155/2019/6051262] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/03/2018] [Revised: 01/07/2019] [Accepted: 01/16/2019] [Indexed: 02/06/2023]
Abstract
Hypoxia-inducible gene domain family member 1A (Higd1a) has recently been reported to protect cells from hypoxia by helping to maintain normal mitochondrial function. The potential induction of Higd1a under high-fat exposure and whether it could protect cells from oxidative stress attracted our attention. Initially, 0.4 mM oleic acid and 0.2 mM palmitate were added to the growth media of HepG2 and LO2 cells for 72 hours. We discovered increased Higd1a expression, and knocking down Higd1a impaired mitochondrial transmembrane potential and induced cell apoptosis. We then identified that elevated reactive oxygen species (ROS) is responsible for increased Higd1a expression. Furthermore, we found that ROS promoted Higd1a expression by upregulating HIF-1a and PGC-1a expressions, and these two proteins could exert synergistic effects in inducing Higd1a expression. Taken together, these data suggest that Higd1a plays positive roles in protecting cells from oxidative stress, and ROS could induce Higd1a expression by upregulating PGC-1a and HIF-1a expressions.
Collapse
|
46
|
Betsinger CN, Cristea IM. Mitochondrial Function, Metabolic Regulation, and Human Disease Viewed through the Prism of Sirtuin 4 (SIRT4) Functions. J Proteome Res 2019; 18:1929-1938. [PMID: 30913880 DOI: 10.1021/acs.jproteome.9b00086] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
As cellular metabolic hubs, mitochondria are the main energy producers for the cell. These organelles host essential energy producing biochemical processes, including the TCA cycle, fatty acid oxidation, and oxidative phosphorylation. An accumulating body of literature has demonstrated that a majority of mitochondrial proteins are decorated with diverse posttranslational modifications (PTMs). Given the critical roles of these proteins in cellular metabolic pathways and response to environmental stress or pathogens, understanding the role of PTMs in regulating their functions has become an area of intense investigation. A major family of enzymes that regulate PTMs within the mitochondria are sirtuins (SIRTs). Albeit until recently the least understood sirtuin, SIRT4 has emerged as an enzyme capable of removing diverse PTMs from its substrates, thereby modulating their functions. SIRT4 was shown to have ADP-ribosyltransferase, deacetylase, lipoamidase, and deacylase enzymatic activities. As metabolic dysfunction is linked to human disease, SIRT4 levels and activities have been implicated in modulating susceptibility to hyperinsulinemia and diabetes, liver disease, cancer, neurodegeneration, heart disease, aging, and pathogenic infections. Therefore, SIRT4 has emerged as a possible candidate for targeted therapeutics. Here, we discuss the diverse enzymatic activities and substrates of SIRT4 and its roles in human health and disease.
Collapse
Affiliation(s)
- Cora N Betsinger
- Department of Molecular Biology , Princeton University , Princeton , New Jersey 08544 , United States
| | - Ileana M Cristea
- Department of Molecular Biology , Princeton University , Princeton , New Jersey 08544 , United States
| |
Collapse
|
47
|
Santaniello S, Cruciani S, Basoli V, Balzano F, Bellu E, Garroni G, Ginesu GC, Cossu ML, Facchin F, Delitala AP, Ventura C, Maioli M. Melatonin and Vitamin D Orchestrate Adipose Derived Stem Cell Fate by Modulating Epigenetic Regulatory Genes. Int J Med Sci 2018; 15:1631-1639. [PMID: 30588186 PMCID: PMC6299418 DOI: 10.7150/ijms.27669] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/05/2018] [Accepted: 08/29/2018] [Indexed: 02/07/2023] Open
Abstract
Melatonin, that regulates many physiological processes including circadian rhythms, is a molecule able to promote osteoblasts maturation in vitro and to prevent bone loss in vivo, while regulating also adipocytes metabolism. In this regard, we have previously shown that melatonin in combination with vitamin D, is able to counteract the appearance of an adipogenic phenotype in adipose derived stem cells (ADSCs), cultured in an adipogenic favoring condition. In the present study, we aimed at evaluating the specific phenotype elicited by melatonin and vitamin D based medium, considering also the involvement of epigenetic regulating genes. ADSCs were cultured in a specific adipogenic conditioned media, in the presence of melatonin alone or with vitamin D. The expression of specific osteogenic related genes was evaluated at different time points, together with the histone deacetylases epigenetic regulators, HDAC1 and Sirtuins (SIRT) 1 and 2. Our results show that melatonin and vitamin D are able to modulate ADSCs commitment towards osteogenic phenotype through the upregulation of HDAC1, SIRT 1 and 2, unfolding an epigenetic regulation in stem cell differentiation and opening novel strategies for future therapeutic balancing of stem cell fate toward adipogenic or osteogenic phenotype.
Collapse
Affiliation(s)
- Sara Santaniello
- Department of Biomedical Sciences, University of Sassari, Viale San Pietro 43/B, 07100 Sassari, Italy
- Laboratory of Molecular Biology and Stem Cell Engineering, National Institute of Biostructures and Biosystems - Eldor Lab, Innovation Accelerator, CNR, Via Piero Gobetti 101, 40129 Bologna, Italy
| | - Sara Cruciani
- Department of Biomedical Sciences, University of Sassari, Viale San Pietro 43/B, 07100 Sassari, Italy
- Laboratory of Molecular Biology and Stem Cell Engineering, National Institute of Biostructures and Biosystems - Eldor Lab, Innovation Accelerator, CNR, Via Piero Gobetti 101, 40129 Bologna, Italy
| | - Valentina Basoli
- Department of Biomedical Sciences, University of Sassari, Viale San Pietro 43/B, 07100 Sassari, Italy
- Laboratory of Molecular Biology and Stem Cell Engineering, National Institute of Biostructures and Biosystems - Eldor Lab, Innovation Accelerator, CNR, Via Piero Gobetti 101, 40129 Bologna, Italy
| | - Francesca Balzano
- Department of Biomedical Sciences, University of Sassari, Viale San Pietro 43/B, 07100 Sassari, Italy
| | - Emanuela Bellu
- Department of Biomedical Sciences, University of Sassari, Viale San Pietro 43/B, 07100 Sassari, Italy
| | - Giuseppe Garroni
- Department of Biomedical Sciences, University of Sassari, Viale San Pietro 43/B, 07100 Sassari, Italy
| | - Giorgio Carlo Ginesu
- General Surgery Unit 2 “Clinica Chirurgica” Medical, Surgical and Experimental Sciences Department, University of Sassari, Viale San Pietro 8, 07100, Sassari, Italy
| | - Maria Laura Cossu
- General Surgery Unit 2 “Clinica Chirurgica” Medical, Surgical and Experimental Sciences Department, University of Sassari, Viale San Pietro 8, 07100, Sassari, Italy
| | - Federica Facchin
- Department of Experimental, Diagnostic and Speciality Medicine (DIMES), University of Bologna, Via Massarenti 9, 40138 Bologna, Italy
| | | | - Carlo Ventura
- Laboratory of Molecular Biology and Stem Cell Engineering, National Institute of Biostructures and Biosystems - Eldor Lab, Innovation Accelerator, CNR, Via Piero Gobetti 101, 40129 Bologna, Italy
| | - Margherita Maioli
- Department of Biomedical Sciences, University of Sassari, Viale San Pietro 43/B, 07100 Sassari, Italy
- Laboratory of Molecular Biology and Stem Cell Engineering, National Institute of Biostructures and Biosystems - Eldor Lab, Innovation Accelerator, CNR, Via Piero Gobetti 101, 40129 Bologna, Italy
- Center for Developmental Biology and Reprogramming- CEDEBIOR, Department of Biomedical Sciences, University of Sassari, Viale San Pietro 43/B, 07100, Sassari, Italy
- Institute of Genetic and Biomedic Research, Consiglio Nazionale delle Ricerche (CNR), Monserrato, Cagliari, Italy
| |
Collapse
|
48
|
Tarantino G, Finelli C, Scopacasa F, Pasanisi F, Contaldo F, Capone D, Savastano S. Corrigendum to "Circulating Levels of Sirtuin 4, a Potential Marker of Oxidative Metabolism, Related to Coronary Artery Disease in Obese Patients Suffering from NAFLD, with Normal or Slightly Increased Liver Enzymes". OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2018; 2018:6357164. [PMID: 30186546 PMCID: PMC6109998 DOI: 10.1155/2018/6357164] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 05/19/2018] [Accepted: 07/11/2018] [Indexed: 02/07/2023]
Abstract
[This corrects the article DOI: 10.1155/2014/920676.].
Collapse
Affiliation(s)
- Giovanni Tarantino
- Department of Clinical Medicine and Surgery, Federico II University Medical School of Naples, Via Sergio Pansini 5, 80131 Naples, Italy
- Centro Ricerche Oncologiche di Mercogliano, Istituto Nazionale Per Lo Studio E La Cura Dei Tumori “Fondazione Giovanni Pascale”, IRCCS, 83013 Mercogliano, Italy
| | | | - Franco Scopacasa
- Department of Biochemistry and Medical Biotechnology, Federico II University Medical School of Naples, Via Sergio Pansini 5, 80131 Naples, Italy
| | - Fabrizio Pasanisi
- Department of Clinical Medicine and Surgery, Federico II University Medical School of Naples, Via Sergio Pansini 5, 80131 Naples, Italy
| | - Franco Contaldo
- Department of Clinical Medicine and Surgery, Federico II University Medical School of Naples, Via Sergio Pansini 5, 80131 Naples, Italy
| | - Domenico Capone
- Department of Neuroscience, Unit of Clinical Pharmacology, Federico II University Medical School of Naples, Via Sergio Pansini 5, 80131 Naples, Italy
| | - Silvia Savastano
- Department of Clinical Medicine and Surgery, Federico II University Medical School of Naples, Via Sergio Pansini 5, 80131 Naples, Italy
| |
Collapse
|
49
|
Simó-Mirabet P, Perera E, Calduch-Giner JA, Afonso JM, Pérez-Sánchez J. Co-expression Analysis of Sirtuins and Related Metabolic Biomarkers in Juveniles of Gilthead Sea Bream ( Sparus aurata) With Differences in Growth Performance. Front Physiol 2018; 9:608. [PMID: 29922168 PMCID: PMC5996159 DOI: 10.3389/fphys.2018.00608] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2018] [Accepted: 05/04/2018] [Indexed: 12/16/2022] Open
Abstract
Sirtuins (SIRTs) represent a conserved protein family of deacetylases that act as master regulators of metabolism, but little is known about their roles in fish and livestock animals in general. The present study aimed to assess the value of SIRTs for the metabolic phenotyping of fish by assessing their co-expression with a wide-representation of markers of energy and lipid metabolism and intestinal function and health in two genetically different gilthead sea bream strains with differences in growth performance. Fish from the fast-growing strain exhibited higher feed intake, feed efficiency and plasma IGF-I levels, along with higher hepatosomatic index and lower mesenteric fat (lean phenotype). These observations suggest differences in tissue energy partitioning with an increased flux of fatty acids from adipose tissue toward the liver. The resulting increased risk of hepatic steatosis may be counteracted in the liver by reduced lipogenesis and enhanced triglyceride catabolism, in combination with a higher and more efficient oxidative metabolism in white skeletal muscle. These effects were supported by co-regulated changes in the expression profile of SIRTs (liver, sirt1; skeletal muscle, sirt2; adipose tissue, sirt5-6) and markers of oxidative metabolism (pgc1α, cpt1a, cs, nd2, cox1), mitochondrial respiration uncoupling (ucp3) and fatty acid and triglyceride metabolism (pparα, pparγ, elovl5, scd1a, lpl, atgl) that were specific to each strain and tissue. The anterior intestine of the fast-growing strain was better suited to cope with improved growth by increased expression of markers of nutrient absorption (fabp2), epithelial barrier integrity (cdh1, cdh17) and immunity (il1β, cd8b, lgals1, lgals8, sIgT, mIgT), which were correlated with low expression levels of sirt4 and markers of fatty acid oxidation (cpt1a). In the posterior intestine, the fast-growing strain showed a consistent up-regulation of sirt2, sirt3, sirt5 and sirt7 concurrently with increased expression levels of markers of cell proliferation (pcna), oxidative metabolism (nd2) and immunity (sIgT, mIgT). Together, these findings indicate that SIRTs may play different roles in the regulation of metabolism, inflammatory tone and growth in farmed fish, arising as powerful biomarkers for a reliable metabolic phenotyping of fish at the tissue-specific level.
Collapse
Affiliation(s)
- Paula Simó-Mirabet
- Nutrigenomics and Fish Growth Endocrinology, Institute of Aquaculture Torre de la Sal-CSIC, Castellón, Spain
| | - Erick Perera
- Nutrigenomics and Fish Growth Endocrinology, Institute of Aquaculture Torre de la Sal-CSIC, Castellón, Spain
| | - Josep A Calduch-Giner
- Nutrigenomics and Fish Growth Endocrinology, Institute of Aquaculture Torre de la Sal-CSIC, Castellón, Spain
| | - Juan M Afonso
- Aquaculture Research Group, Institute of Sustainable Aquaculture and Marine Ecosystems (IU-ECOAQUA), University of Las Palmas de Gran Canaria (GIA), Las Palmas, Spain
| | - Jaume Pérez-Sánchez
- Nutrigenomics and Fish Growth Endocrinology, Institute of Aquaculture Torre de la Sal-CSIC, Castellón, Spain
| |
Collapse
|
50
|
Singh CK, Chhabra G, Ndiaye MA, Garcia-Peterson LM, Mack NJ, Ahmad N. The Role of Sirtuins in Antioxidant and Redox Signaling. Antioxid Redox Signal 2018; 28:643-661. [PMID: 28891317 PMCID: PMC5824489 DOI: 10.1089/ars.2017.7290] [Citation(s) in RCA: 547] [Impact Index Per Article: 78.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
SIGNIFICANCE Antioxidant and redox signaling (ARS) events are regulated by critical molecules that modulate antioxidants, reactive oxygen species (ROS) or reactive nitrogen species (RNS), and/or oxidative stress within the cell. Imbalances in these molecules can disturb cellular functions to become pathogenic. Sirtuins serve as important regulators of ARS in cells. Recent Advances: Sirtuins (SIRTs 1-7) are a family of nicotinamide adenine dinucleotide (NAD)-dependent histone deacetylases with the ability to deacetylate histone and nonhistone targets. Recent studies show that sirtuins modulate the regulation of a variety of cellular processes associated with ARS. SIRT1, SIRT3, and SIRT5 protect the cell from ROS, and SIRT2, SIRT6, and SIRT7 modulate key oxidative stress genes and mechanisms. Interestingly, SIRT4 has been shown to induce ROS production and has antioxidative roles as well. CRITICAL ISSUES A complete understanding of the roles of sirtuins in redox homeostasis of the cell is very important to understand the normal functioning as well as pathological manifestations. In this review, we have provided a critical discussion on the role of sirtuins in the regulation of ARS. We have also discussed mechanistic interactions among different sirtuins. Indeed, a complete understanding of sirtuin biology could be critical at multiple fronts. FUTURE DIRECTIONS Sirtuins are emerging to be important in normal mammalian physiology and in a variety of oxidative stress-mediated pathological situations. Studies are needed to dissect the mechanisms of sirtuins in maintaining redox homeostasis. Efforts are also required to assess the targetability of sirtuins in the management of redox-regulated diseases. Antioxid. Redox Signal. 28, 643-661.
Collapse
Affiliation(s)
- Chandra K Singh
- Department of Dermatology, University of Wisconsin , Madison, Wisconsin
| | - Gagan Chhabra
- Department of Dermatology, University of Wisconsin , Madison, Wisconsin
| | - Mary Ann Ndiaye
- Department of Dermatology, University of Wisconsin , Madison, Wisconsin
| | | | - Nicholas J Mack
- Department of Dermatology, University of Wisconsin , Madison, Wisconsin
| | - Nihal Ahmad
- Department of Dermatology, University of Wisconsin , Madison, Wisconsin
| |
Collapse
|