1
|
Zou Y, Tian L, Pei L, Hao J, Chen T, Qi J, Qiu J, Xu Y, Hu X, Chen L, Dou X. SFAs facilitates ceramide's de novo synthesis via TLR4 and intensifies hepatocyte lipotoxicity. Int Immunopharmacol 2025; 147:114020. [PMID: 39793229 DOI: 10.1016/j.intimp.2025.114020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2024] [Revised: 12/18/2024] [Accepted: 01/02/2025] [Indexed: 01/13/2025]
Abstract
BACKGROUND Non-alcoholic steatohepatitis (NASH), an advanced manifestation of non-alcoholic fatty liver disease (NAFLD), is characterized by hepatocyte injury, inflammation, and fibrosis. Saturated fatty acids (SFAs) have emerged as key contributors to hepatocyte lipotoxicity and disease progression. Toll-like receptor 4 (TLR4) acts as a sentinel for diverse ligands, including lipopolysaccharide (LPS) and endogenous molecules like palmitic acid (PA)-induced ceramide (CER) accumulation, promoting hepatocyte demise. However, the intricate mechanisms underlying TLR4's modulation of ceramide metabolism and their concerted effect on SFA-mediated hepatotoxicity remain elusive. METHODS A NASH mouse model with liver-specific TLR4 knockdown was established through palm oil feeding and AAV2/8 tail vein injection. Histological and biochemical assessments were conducted to evaluate the mice's condition and liver damage extent. Liquid chromatography-mass spectrometry (LC-MS) was employed to quantify ceramide levels in liver tissues, offering insights into NASH mechanisms. RESULTS The PO-fed model exhibited elevated serum ALT, AST, and liver TG levels, enhancing lipid accumulation and hepatocellular damage. TLR4 knock-down reduced liver mass and the liver-to-body weight ratio, signifying a decreased hepatic burden. Histopathological evaluations revealed substantial improvement in hepatic steatosis in TLR4-silenced PO-fed mice, with diminished lipid droplets and inflammatory infiltrates. LC-MS analysis showed a marked decrease in long-chain ceramides (C14, C16, C20) in TLR4-knockdown PO-fed mice. Furthermore, expression of MyD88, SPTLC1, SPTLC2, and inflammatory markers IL-1β, IL-6, TNF-α were significantly attenuated. CONCLUSION SFAs activate the TLR4 signaling pathway via MyD88, fostering ceramide de novo synthesis, which exacerbates hepatocyte lipotoxicity and accelerates NASH progression.
Collapse
Affiliation(s)
- Yuchao Zou
- School of Life Science, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, 310053, PR China
| | - Lulu Tian
- School of Life Science, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, 310053, PR China
| | - Liuhua Pei
- School of Public Health, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, 310053, PR China
| | - Jie Hao
- School of Life Science, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, 310053, PR China
| | - Tianhang Chen
- School of Life Science, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, 310053, PR China
| | - Jiayu Qi
- School of Life Science, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, 310053, PR China
| | - Jiannan Qiu
- School of Life Science, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, 310053, PR China
| | - Yinuo Xu
- School of Life Science, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, 310053, PR China
| | - Xiaokai Hu
- School of Life Science, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, 310053, PR China
| | - Lin Chen
- School of Life Science, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, 310053, PR China.
| | - Xiaobing Dou
- School of Life Science, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, 310053, PR China.
| |
Collapse
|
2
|
Kuriakose BB, Zwamel AH, Mutar AA, Uthirapathy S, Bishoyi AK, Naidu KS, Hjazi A, Nakash P, Arya R, Almalki SG. The critical role of NLRP3 in drug resistance of cancers: Focus on the molecular mechanisms and possible therapeutics. Semin Oncol 2025; 52:27-40. [PMID: 40037148 DOI: 10.1016/j.seminoncol.2025.152337] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/24/2024] [Revised: 02/08/2025] [Accepted: 02/12/2025] [Indexed: 03/06/2025]
Abstract
Nod-like receptor protein 3 (NLRP3) is a member of the leucine-rich repeat-containing protein (NLR) canonical inflammasome family. It regulates the pathophysiology of cancer by facilitating immune responses and apoptotic proteins. Furthermore, it has been observed that chemotherapy activates NLRP3 in human malignancies. The secretion of IL-1β and IL-22 to promote cancer spread may be triggered by NLRP3 activation. Furthermore, earlier studies have exhibited that NLRP3 may cause medication resistance when used in cancer treatments given that cell viability may be regulated by NLRP3 depletion. Additionally, clinical studies have demonstrated correlation between NLRP3 expression, lymphogenesis, and cancer metastasis. Various NLRP3 agonists may cause the EMT process, stimulate IL-1β and Wnt/β-catenin signaling, and alter miRNA function in drug-resistant cells. This review seeks to clarify the possibility involvement of NLRP3-related pathways in the control of cancer cells' resistance to widely used treatment approaches, such as chemotherapy. In the end, an improved perception of the corresponding mechanisms behind NLRP3's tumor-supporting activities will help NLRP3-based treatments advance in the future.
Collapse
Affiliation(s)
- Beena Briget Kuriakose
- Department of Basic Medical Sciences, College of Applied Medical Sciences, King khalid University, Khamis Mushayt, Kingdom of Saudi Arabia
| | - Ahmed Hussein Zwamel
- Department of medical analysis, Medical laboratory technique college, the Islamic University, Najaf, Iraq; Department of medical analysis, Medical laboratory technique college, the Islamic University of Al Diwaniyah, Al Diwaniyah, Iraq; Department of medical analysis, Medical laboratory technique college, the Islamic University of Babylon, Babylon, Iraq
| | - Ayad Abdulrazzaq Mutar
- Medical Laboratory Techniques department, College of Health and medical technology, Al-maarif University, Anbar, Iraq.
| | - Subasini Uthirapathy
- Pharmacy Department, Tishk International University, Erbil, Kurdistan Region, Iraq
| | - Ashok Kumar Bishoyi
- Marwadi University Research Center, Department of Microbiology, Faculty of Science, Marwadi University, Rajkot, Gujarat, India
| | - K Satyam Naidu
- Department of Chemistry, Raghu Engineering College, Visakhapatnam, Andhra Pradesh, India
| | - Ahmed Hjazi
- Department of Medical Laboratory, College of Applied Medical Sciences, Princse Sattam bin Abdulaziz University, Al-Kharj, Saudi Arabia
| | - Prashant Nakash
- NIMS Institute of Pharmacy, NIMS University Rajasthan, Jaipur, Rajasthan, India
| | - Renu Arya
- Chandigarh Pharmacy College, Chandigarh Group of Colleges-Jhanjeri, Mohali, Punjab, India
| | - Sami G Almalki
- Department of Medical Laboratory Sciences, College of Applied Medical Sciences, Majmaah University, Majmaah, Saudi Arabia
| |
Collapse
|
3
|
Pandey A, Li Z, Gautam M, Ghosh A, Man SM. Molecular mechanisms of emerging inflammasome complexes and their activation and signaling in inflammation and pyroptosis. Immunol Rev 2025; 329:e13406. [PMID: 39351983 PMCID: PMC11742652 DOI: 10.1111/imr.13406] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2025]
Abstract
Inflammasomes are multi-protein complexes that assemble within the cytoplasm of mammalian cells in response to pathogen-associated molecular patterns (PAMPs) or damage-associated molecular patterns (DAMPs), driving the secretion of the pro-inflammatory cytokines IL-1β and IL-18, and pyroptosis. The best-characterized inflammasome complexes are the NLRP3, NAIP-NLRC4, NLRP1, AIM2, and Pyrin canonical caspase-1-containing inflammasomes, and the caspase-11 non-canonical inflammasome. Newer inflammasome sensor proteins have been identified, including NLRP6, NLRP7, NLRP9, NLRP10, NLRP11, NLRP12, CARD8, and MxA. These inflammasome sensors can sense PAMPs from bacteria, viruses and protozoa, or DAMPs in the form of mitochondrial damage, ROS, stress and heme. The mechanisms of action, physiological relevance, consequences in human diseases, and avenues for therapeutic intervention for these novel inflammasomes are beginning to be realized. Here, we discuss these emerging inflammasome complexes and their putative activation mechanisms, molecular and signaling pathways, and physiological roles in health and disease.
Collapse
Affiliation(s)
- Abhimanu Pandey
- Division of Immunology and Infectious Diseases, The John Curtin School of Medical ResearchThe Australian National UniversityCanberraAustralia
| | - Zheyi Li
- Division of Immunology and Infectious Diseases, The John Curtin School of Medical ResearchThe Australian National UniversityCanberraAustralia
| | - Manjul Gautam
- Division of Immunology and Infectious Diseases, The John Curtin School of Medical ResearchThe Australian National UniversityCanberraAustralia
| | - Aritra Ghosh
- Division of Immunology and Infectious Diseases, The John Curtin School of Medical ResearchThe Australian National UniversityCanberraAustralia
| | - Si Ming Man
- Division of Immunology and Infectious Diseases, The John Curtin School of Medical ResearchThe Australian National UniversityCanberraAustralia
| |
Collapse
|
4
|
Anand PK. From fat to fire: The lipid-inflammasome connection. Immunol Rev 2025; 329:e13403. [PMID: 39327931 PMCID: PMC11744241 DOI: 10.1111/imr.13403] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/28/2024]
Abstract
Inflammasomes are multiprotein complexes that play a crucial role in regulating immune responses by governing the activation of Caspase-1, the secretion of pro-inflammatory cytokines, and the induction of inflammatory cell death, pyroptosis. The inflammasomes are pivotal in effective host defense against a range of pathogens. Yet, overt activation of inflammasome signaling can be detrimental. The most well-studied NLRP3 inflammasome has the ability to detect a variety of stimuli including pathogen-associated molecular patterns, environmental irritants, and endogenous stimuli released from dying cells. Additionally, NLRP3 acts as a key sensor of cellular homeostasis and can be activated by disturbances in diverse metabolic pathways. Consequently, NLRP3 is considered a key player linking metabolic dysregulation to numerous inflammatory disorders such as gout, diabetes, and atherosclerosis. Recently, compelling studies have highlighted a connection between lipids and the regulation of NLRP3 inflammasome. Lipids are integral to cellular processes that serve not only in maintaining the structural integrity and subcellular compartmentalization, but also in contributing to physiological equilibrium. Certain lipid species are known to define NLRP3 subcellular localization, therefore directly influencing the site of inflammasome assembly and activation. For instance, phosphatidylinositol 4-phosphate plays a crucial role in NLRP3 localization to the trans Golgi network. Moreover, new evidence has demonstrated the roles of lipid biosynthesis and trafficking in activation of the NLRP3 inflammasome. This review summarizes and discusses these emerging and varied roles of lipid metabolism in inflammasome activation. A deeper understanding of lipid-inflammasome interactions may open new avenues for therapeutic interventions to prevent or treat chronic inflammatory and autoimmune conditions.
Collapse
Affiliation(s)
- Paras K. Anand
- Department of Infectious Disease, Faculty of MedicineImperial College LondonLondonUK
| |
Collapse
|
5
|
Wu J, Shyy M, Shyy JYJ, Xiao H. Role of inflammasomes in endothelial dysfunction. Br J Pharmacol 2024; 181:4958-4972. [PMID: 38952037 DOI: 10.1111/bph.16479] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2023] [Revised: 03/14/2024] [Accepted: 05/04/2024] [Indexed: 07/03/2024] Open
Abstract
The vascular endothelium dynamically responds to environmental cues and plays a pivotal role in maintaining vascular homeostasis by regulating vasomotor tone, blood cell trafficking, permeability and immune responses. However, endothelial dysfunction results in various pathological conditions. Inflammasomes are large intracellular multimeric complexes activated by pathogens or cellular damage. Inflammasomes in vascular endothelial cells (ECs) initiate innate immune responses, which have emerged as significant mediators in endothelial dysfunction, contributing to the pathophysiology of an array of diseases. This review summarizes the mechanisms and ramifications of inflammasomes in ECs and related vascular diseases such as atherosclerosis, abdominal aortic aneurysm, stroke, and lung and kidney diseases. We also discuss potential drugs targeting EC inflammasomes and their applications in treating vascular diseases.
Collapse
Affiliation(s)
- Jimin Wu
- Department of Cardiology and Institute of Vascular Medicine, Peking University Third Hospital, Beijing, China
- State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, China
- Beijing Key Laboratory of Cardiovascular Receptors Research, Beijing, China
- Research Unit of Medical Science Research Management/Basic and Clinical Research of Metabolic Cardiovascular Diseases, Chinese Academy of Medical Sciences, Beijing, China
- Haihe Laboratory of Cell Ecosystem, Beijing, China
| | - Melody Shyy
- Biological Sciences, University of California, Santa Barbara, Santa Barbara, California, USA
| | - John Y-J Shyy
- Division of Cardiology, Department of Medicine, University of California, San Diego, La Jolla, California, USA
| | - Han Xiao
- Department of Cardiology and Institute of Vascular Medicine, Peking University Third Hospital, Beijing, China
- State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, China
- Beijing Key Laboratory of Cardiovascular Receptors Research, Beijing, China
- Research Unit of Medical Science Research Management/Basic and Clinical Research of Metabolic Cardiovascular Diseases, Chinese Academy of Medical Sciences, Beijing, China
- Haihe Laboratory of Cell Ecosystem, Beijing, China
| |
Collapse
|
6
|
Wen Y, Yi F, Zhang J, Wang Y, Zhao C, Zhao B, Wang J. Uncovering the protective mechanism of baicalin in treatment of fatty liver based on network pharmacology and cell model of NAFLD. Int Immunopharmacol 2024; 141:112954. [PMID: 39153306 DOI: 10.1016/j.intimp.2024.112954] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Revised: 07/25/2024] [Accepted: 08/13/2024] [Indexed: 08/19/2024]
Abstract
Excessive nonesterified fatty acids (NEFA) impair cellular metabolism and will induce fatty liver formation in dairy cows during the periparturient. Baicalin, an active flavonoid, has great potential efficacy in alleviating lipid accumulation and ameliorating the development of fatty liver disease. Nevertheless, its mechanism remains unclear. Here, the potential mechanism of baicalin on system levels was explored using network pharmacology and in vitro experiments. Firstly, the target of baicalin and fatty liver disease was predicted, and then the protein-protein interaction (PPI) network was constructed. In addition, the Kyoto Encyclopedia of Genes and Genomes (KEGG) (q-value) pathway enrichment is performed through the Database for Annotation, Visualization, and Integrated Discovery (DAVID) server. Finally, the results of the network analysis of the in vitro treatment of bovine hepatocytes by NEFA were confirmed. The results showed that 33 relevant targets of baicalin in the treatment of liver fatty were predicted by network pharmacology, and the top 20 relevant pathways were extracted by KEGG database. Baicalin treatment can reduce triglyceride (TAG) content and lipid droplet accumulation in NEFA-treated bovine hepatocytes, and the mechanism is related to inhibiting lipid synthesis and promoting lipid oxidation. The alleviating effect of baicalin on fatty liver may be related to the up-regulation of solute vector family member 4 (SLC2A4), Down-regulated AKT serine/threonine kinase 1 (AKT1), Peroxisome proliferator-activated receptor gamma (PPARG), Epidermal growth factor receptor (EGFR), tumor necrosis factor (TNF), Interleukin 6 (IL-6) were associated. These results suggested that baicalin may modulate key inflammatory markers, and lipogenesis processes to prevent fatty liver development in dairy cows.
Collapse
Affiliation(s)
- Yongqiang Wen
- College of Veterinary Medicine, Northwest A&F University, Yangling 712100, China
| | - Fanxuan Yi
- College of Veterinary Medicine, Northwest A&F University, Yangling 712100, China
| | - Jia Zhang
- College of Veterinary Medicine, Northwest A&F University, Yangling 712100, China
| | - Yazhou Wang
- College of Veterinary Medicine, Northwest A&F University, Yangling 712100, China
| | - Chenxu Zhao
- College of Veterinary Medicine, Northwest A&F University, Yangling 712100, China
| | - Baoyu Zhao
- College of Veterinary Medicine, Northwest A&F University, Yangling 712100, China
| | - Jianguo Wang
- College of Veterinary Medicine, Northwest A&F University, Yangling 712100, China.
| |
Collapse
|
7
|
Haghighi E, Abolmaali SS, Dehshahri A, Mousavi Shaegh SA, Azarpira N, Tamaddon AM. Navigating the intricate in-vivo journey of lipid nanoparticles tailored for the targeted delivery of RNA therapeutics: a quality-by-design approach. J Nanobiotechnology 2024; 22:710. [PMID: 39543630 PMCID: PMC11566655 DOI: 10.1186/s12951-024-02972-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Accepted: 11/03/2024] [Indexed: 11/17/2024] Open
Abstract
RNA therapeutics, such as mRNA, siRNA, and CRISPR-Cas9, present exciting avenues for treating diverse diseases. However, their potential is commonly hindered by vulnerability to degradation and poor cellular uptake, requiring effective delivery systems. Lipid nanoparticles (LNPs) have emerged as a leading choice for in vivo RNA delivery, offering protection against degradation, enhanced cellular uptake, and facilitation of endosomal escape. However, LNPs encounter numerous challenges for targeted RNA delivery in vivo, demanding advanced particle engineering, surface functionalization with targeting ligands, and a profound comprehension of the biological milieu in which they function. This review explores the structural and physicochemical characteristics of LNPs, in-vivo fate, and customization for RNA therapeutics. We highlight the quality-by-design (QbD) approach for targeted delivery beyond the liver, focusing on biodistribution, immunogenicity, and toxicity. In addition, we explored the current challenges and strategies associated with LNPs for in-vivo RNA delivery, such as ensuring repeated-dose efficacy, safety, and tissue-specific gene delivery. Furthermore, we provide insights into the current clinical applications in various classes of diseases and finally prospects of LNPs in RNA therapeutics.
Collapse
Affiliation(s)
- Elahe Haghighi
- Department of Pharmaceutical Nanotechnology, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Samira Sadat Abolmaali
- Department of Pharmaceutical Nanotechnology, Shiraz University of Medical Sciences, Shiraz, Iran.
- Center for Nanotechnology in Drug Delivery, Shiraz University of Medical Sciences, Shiraz, Iran.
| | - Ali Dehshahri
- Center for Nanotechnology in Drug Delivery, Shiraz University of Medical Sciences, Shiraz, Iran
- Department of Pharmaceutical Biotechnology, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Seyed Ali Mousavi Shaegh
- Laboratory of Microfluidics and Medical Microsystems, Research Institute for Medical Sciences, Mashhad University of Medical Sciences, Mashhad, Iran
- Orthopedic Research Center, Ghaem Hospital, Mashhad University of Medical Sciences, Mashhad, Iran
- Clinical Research Development Unit, Ghaem Hospital, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Negar Azarpira
- Stem Cells Technology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Ali Mohammad Tamaddon
- Department of Pharmaceutical Nanotechnology, Shiraz University of Medical Sciences, Shiraz, Iran.
- Center for Nanotechnology in Drug Delivery, Shiraz University of Medical Sciences, Shiraz, Iran.
- Department of Pharmaceutics, Shiraz University of Medical Sciences, Shiraz, Iran.
| |
Collapse
|
8
|
Szukiewicz D. Insights into Reproductive Immunology and Placental Pathology. Int J Mol Sci 2024; 25:12135. [PMID: 39596208 PMCID: PMC11594814 DOI: 10.3390/ijms252212135] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2024] [Revised: 11/09/2024] [Accepted: 11/10/2024] [Indexed: 11/28/2024] Open
Abstract
The formation of a daughter organism as a result of the fusion of an egg and a sperm cell, followed by the implantation of the embryo, the formation of the placenta, and the further growth of the embryo and then fetus until delivery, poses particular challenges for the immune system [...].
Collapse
Affiliation(s)
- Dariusz Szukiewicz
- Department of Biophysics, Physiology & Pathophysiology, Faculty of Health Sciences, Medical University of Warsaw, 02-004 Warsaw, Poland
| |
Collapse
|
9
|
Fang Y, Liu X, Ren J, Wang X, Zhou F, Huang S, You L, Zhao Y. Integrated analysis of microbiome and metabolome reveals signatures in PDAC tumorigenesis and prognosis. Microbiol Spectr 2024; 12:e0096224. [PMID: 39387592 PMCID: PMC11540152 DOI: 10.1128/spectrum.00962-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Accepted: 09/16/2024] [Indexed: 10/15/2024] Open
Abstract
Pancreatic cancer, predominantly pancreatic ductal adenocarcinoma (PDAC), is one of the most malignant tumors of the digestive system. Emerging evidence suggests the involvement of the microbiome and metabolic substances in the development of PDAC, yet the results remain contradictory. This study aims to identify the alterations and relationships in intratumoral microbiome and metabolites in PDAC. We collected matched tumor and normal adjacent tissue (NAT) samples from 105 PDAC patients and performed a 6-year follow-up. 2bRAD-M sequencing, untargeted liquid chromatography-tandem mass spectrometry, and untargeted gas chromatography-mass spectrometry were performed. Compared with NATs, microbial α-diversity decreased in PDAC tumors. The relative abundance of Staphylococcus aureus, Cutibacterium acnes, and Cutibacterium granulosum was higher in PDAC tumor after adjusting for confounding factors body mass index and M stage, and the presence of Ralstonia pickettii_B was found associated with a worse overall survival. Metabolomic analysis revealed distinctive differences in composition between PDAC and NAT, with 553 discriminative metabolites identified. Differential metabolites were revealed to originate from the microbiota and showed significant interactions with shifted bacterial species through KO (KEGG Orthology) genes. These findings suggest that the PDAC microenvironment harbors unique microbial-derived enzymatic reactions, potentially influencing the occurrence and development of PDAC by modulating the levels of glycerol-3-phosphate, succinate, carbonate, and beta-alanine. IMPORTANCE We conducted a large sample-size pancreatic adenocarcinoma microbiome study using a novel microbiome sequencing method and two metabolomic assays. Two significant outcomes of our analysis are: (i) commensal opportunistic pathogens Staphylococcus aureus, Cutibacterium acnes, and Cutibacterium granulosum were enriched in pancreatic ductal adenocarcinoma (PDAC) tumors compared with normal adjacent tissues, and (ii) worse overall survival was found related to the presence of Ralstonia pickettii_B. Microbial species affect the tumorigenesis, metastasis, and prognosis of PDAC via unique microbe-enzyme-metabolite interaction. Thus, our study highlights the need for further investigation of the potential associations between pancreatic microbiota-derived omics signatures, which may drive the clinical transformation of microbiome-derived strategies toward therapy-targeted bacteria.
Collapse
Affiliation(s)
- Yuan Fang
- Department of General
Surgery, Peking Union Medical College Hospital, Peking Union Medical
College, Chinese Academy of Medical
Sciences, Beijing,
China
- Key Laboratory of
Research in Pancreatic Tumor, Chinese Academy of Medical
Sciences, Beijing,
China
- National Science and
Technology Key Infrastructure on Translational Medicine in Peking Union
Medical College Hospital,
Beijing, China
- State Key Laboratory
of Complex Severe and Rare Diseases, Peking Union Medical College
Hospital, Chinese Academy of Medical Sciences and Peking Union Medical
College, Beijing,
China
| | - Xiaohong Liu
- Department of General
Surgery, Peking Union Medical College Hospital, Peking Union Medical
College, Chinese Academy of Medical
Sciences, Beijing,
China
- Key Laboratory of
Research in Pancreatic Tumor, Chinese Academy of Medical
Sciences, Beijing,
China
- National Science and
Technology Key Infrastructure on Translational Medicine in Peking Union
Medical College Hospital,
Beijing, China
- State Key Laboratory
of Complex Severe and Rare Diseases, Peking Union Medical College
Hospital, Chinese Academy of Medical Sciences and Peking Union Medical
College, Beijing,
China
| | - Jie Ren
- Department of General
Surgery, Peking Union Medical College Hospital, Peking Union Medical
College, Chinese Academy of Medical
Sciences, Beijing,
China
- Key Laboratory of
Research in Pancreatic Tumor, Chinese Academy of Medical
Sciences, Beijing,
China
- National Science and
Technology Key Infrastructure on Translational Medicine in Peking Union
Medical College Hospital,
Beijing, China
- State Key Laboratory
of Complex Severe and Rare Diseases, Peking Union Medical College
Hospital, Chinese Academy of Medical Sciences and Peking Union Medical
College, Beijing,
China
| | - Xing Wang
- Department of General
Surgery, Peking Union Medical College Hospital, Peking Union Medical
College, Chinese Academy of Medical
Sciences, Beijing,
China
- Key Laboratory of
Research in Pancreatic Tumor, Chinese Academy of Medical
Sciences, Beijing,
China
- National Science and
Technology Key Infrastructure on Translational Medicine in Peking Union
Medical College Hospital,
Beijing, China
- State Key Laboratory
of Complex Severe and Rare Diseases, Peking Union Medical College
Hospital, Chinese Academy of Medical Sciences and Peking Union Medical
College, Beijing,
China
| | - Feihan Zhou
- Department of General
Surgery, Peking Union Medical College Hospital, Peking Union Medical
College, Chinese Academy of Medical
Sciences, Beijing,
China
- Key Laboratory of
Research in Pancreatic Tumor, Chinese Academy of Medical
Sciences, Beijing,
China
- National Science and
Technology Key Infrastructure on Translational Medicine in Peking Union
Medical College Hospital,
Beijing, China
- State Key Laboratory
of Complex Severe and Rare Diseases, Peking Union Medical College
Hospital, Chinese Academy of Medical Sciences and Peking Union Medical
College, Beijing,
China
| | - Shi Huang
- Faculty of Dentistry,
The University of Hong Kong, Hong
Kong SAR, China
| | - Lei You
- Department of General
Surgery, Peking Union Medical College Hospital, Peking Union Medical
College, Chinese Academy of Medical
Sciences, Beijing,
China
- Key Laboratory of
Research in Pancreatic Tumor, Chinese Academy of Medical
Sciences, Beijing,
China
- National Science and
Technology Key Infrastructure on Translational Medicine in Peking Union
Medical College Hospital,
Beijing, China
- State Key Laboratory
of Complex Severe and Rare Diseases, Peking Union Medical College
Hospital, Chinese Academy of Medical Sciences and Peking Union Medical
College, Beijing,
China
| | - Yupei Zhao
- Department of General
Surgery, Peking Union Medical College Hospital, Peking Union Medical
College, Chinese Academy of Medical
Sciences, Beijing,
China
- Key Laboratory of
Research in Pancreatic Tumor, Chinese Academy of Medical
Sciences, Beijing,
China
- National Science and
Technology Key Infrastructure on Translational Medicine in Peking Union
Medical College Hospital,
Beijing, China
- State Key Laboratory
of Complex Severe and Rare Diseases, Peking Union Medical College
Hospital, Chinese Academy of Medical Sciences and Peking Union Medical
College, Beijing,
China
| |
Collapse
|
10
|
Atalay Ekiner S, Gęgotek A, Skrzydlewska E. Inflammasome activity regulation by PUFA metabolites. Front Immunol 2024; 15:1452749. [PMID: 39290706 PMCID: PMC11405227 DOI: 10.3389/fimmu.2024.1452749] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Accepted: 08/19/2024] [Indexed: 09/19/2024] Open
Abstract
Oxidative stress and the accompanying chronic inflammation constitute an important metabolic problem that may lead to pathology, especially when the body is exposed to physicochemical and biological factors, including UV radiation, pathogens, drugs, as well as endogenous metabolic disorders. The cellular response is associated, among others, with changes in lipid metabolism, mainly due to the oxidation and the action of lipolytic enzymes. Products of oxidative fragmentation/cyclization of polyunsaturated fatty acids (PUFAs) [4-HNE, MDA, 8-isoprostanes, neuroprostanes] and eicosanoids generated as a result of the enzymatic metabolism of PUFAs significantly modify cellular metabolism, including inflammation and the functioning of the immune system by interfering with intracellular molecular signaling. The key regulators of inflammation, the effectiveness of which can be regulated by interacting with the products of lipid metabolism under oxidative stress, are inflammasome complexes. An example is both negative or positive regulation of NLRP3 inflammasome activity by 4-HNE depending on the severity of oxidative stress. 4-HNE modifies NLRP3 activity by both direct interaction with NLRP3 and alteration of NF-κB signaling. Furthermore, prostaglandin E2 is known to be positively correlated with both NLRP3 and NLRC4 activity, while its potential interference with AIM2 or NLRP1 activity is unproven. Therefore, the influence of PUFA metabolites on the activity of well-characterized inflammasome complexes is reviewed.
Collapse
Affiliation(s)
| | - Agnieszka Gęgotek
- Department of Analytical Chemistry, Medical University of Bialystok, Bialystok, Poland
| | - Elżbieta Skrzydlewska
- Department of Analytical Chemistry, Medical University of Bialystok, Bialystok, Poland
| |
Collapse
|
11
|
Leishman S, Aljadeed NM, Qian L, Cockcroft S, Behmoaras J, Anand PK. Fatty acid synthesis promotes inflammasome activation through NLRP3 palmitoylation. Cell Rep 2024; 43:114516. [PMID: 39024103 DOI: 10.1016/j.celrep.2024.114516] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Revised: 05/31/2024] [Accepted: 07/01/2024] [Indexed: 07/20/2024] Open
Abstract
Despite its significance, the role of lipid metabolism in NLRP3 inflammasome remains elusive. Here, we reveal a critical role for fatty acid synthase (FASN) in NLRP3 inflammasome activation. We demonstrate that pharmacological or genetic depletion of FASN dampens NLRP3 activation in primary mouse and human macrophages and in mice. This disruption in NLRP3 activation is contingent upon FASN activity. Accordingly, abolishing cellular palmitoylation, a post-translational modification in which the FASN product palmitate is reversibly conjugated to cysteine residues of target proteins, blunts inflammasome signaling. Correspondingly, an acyl-biotin exchange assay corroborated NLRP3 palmitoylation. Mechanistically, Toll-like receptor (TLR) ligation introduces palmitoylation at NLRP3 Cys898, permitting NLRP3 translocation to dispersed trans-Golgi network (dTGN) vesicles, the site of inflammasome assembly, upon NLRP3 activation. Accordingly, the NLRP3 Cys898 mutant exhibits reduced palmitoylation, limited translocation to the dTGN compartment, and diminished inflammasome activation. These results underscore mechanistic insights through which lipid metabolism licenses NLRP3 inflammasome assembly and activation.
Collapse
Affiliation(s)
- Stuart Leishman
- Department of Infectious Disease, Imperial College London, London W12 0NN, UK
| | - Najd M Aljadeed
- Department of Infectious Disease, Imperial College London, London W12 0NN, UK
| | - Liyunhe Qian
- Department of Infectious Disease, Imperial College London, London W12 0NN, UK
| | - Shamshad Cockcroft
- Department of Neuroscience, Physiology and Pharmacology, Division of Biosciences, University College London, London WC1E 6JJ, UK
| | - Jacques Behmoaras
- Programme in Cardiovascular and Metabolic Disorders and Centre for Computational Biology, Duke-NUS Medical School Singapore, Singapore
| | - Paras K Anand
- Department of Infectious Disease, Imperial College London, London W12 0NN, UK.
| |
Collapse
|
12
|
Li Y, He Q, Chen S, Dli H, Zhao J, Sun X, Yang P, Mao Q, Xia H. BI-7273, a BRD9 inhibitor, reduces lipid accumulation by downregulating the AKT/mTOR/SREBP1 signaling pathway. Biochem Pharmacol 2024; 226:116412. [PMID: 38971334 DOI: 10.1016/j.bcp.2024.116412] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2023] [Revised: 04/24/2024] [Accepted: 07/03/2024] [Indexed: 07/08/2024]
Abstract
Increases in de novo lipogenesis that disturbed lipid homeostasis and caused lipid accumulation are a major cause of NAFLD and obesity. SREBP1 is a crucial regulatory factor controlling the expression of rate-limiting enzymes of lipid synthesis. A reduction in SREBP1expression can reduce lipid accumulation. Thus, we utilized an SREBP1-luciferase-KI HEK293 cell line constructed by our lab to screen 200 kinds of epigenetic drugs for their ability to downregulate SREBP1expression. BI-7273, an inhibitor of bromodomain-containing protein 9 (BRD9), was screened and found to decrease SREBP1 expression. What is more, BI-7273 has been confirmed that it could reduce lipid accumulation in HepG2 cells by BODIPY staining, and significantly decrease the protein expression of SREBP1 and FASN. To explore the potential mechanism BI-7273 reducing lipid accumulation, RNA sequencing (RNA-seq) was performed and demonstrated that BI-7273 reduced lipid accumulation by downregulating the AKT/mTOR/SREBP1 pathway in vitro. Finally, these results were verified in NAFLD and obesity mouse model induced by high fat diet (HFD). The results indicated that BI-7273 could decrease mouse body weight and improve insulin sensitivity, but also exhibited a strong negative correlation with serum lipid levels, and also demonstrated that BI-7273 reduced lipid accumulation via AKT/mTOR/SREBP1 pathway in vivo. In conclusion, our results revealed that BI-7273 decreases lipid accumulation by downregulating the AKT/mTOR/SREBP1 pathway in vivo and in vitro. This is the first report demonstrating the protective effect of this BRD9 inhibitor against NAFLD and obesity. BRD9 may be a novel target for the discovery of effective drugs to treat lipid metabolism disorders.
Collapse
Affiliation(s)
- Yu Li
- Laboratory of Gene Therapy, Department of Biochemistry, College of Life Sciences, Shaanxi Normal University, 199 South Chang'an Road, Xi'an 710062, Shaanxi, PR China
| | - Qiongyan He
- Laboratory of Gene Therapy, Department of Biochemistry, College of Life Sciences, Shaanxi Normal University, 199 South Chang'an Road, Xi'an 710062, Shaanxi, PR China
| | - Shuyu Chen
- Laboratory of Gene Therapy, Department of Biochemistry, College of Life Sciences, Shaanxi Normal University, 199 South Chang'an Road, Xi'an 710062, Shaanxi, PR China
| | - Huma Dli
- Laboratory of Gene Therapy, Department of Biochemistry, College of Life Sciences, Shaanxi Normal University, 199 South Chang'an Road, Xi'an 710062, Shaanxi, PR China
| | - Junli Zhao
- Laboratory of Gene Therapy, Department of Biochemistry, College of Life Sciences, Shaanxi Normal University, 199 South Chang'an Road, Xi'an 710062, Shaanxi, PR China
| | - Xiaohong Sun
- Laboratory of Gene Therapy, Department of Biochemistry, College of Life Sciences, Shaanxi Normal University, 199 South Chang'an Road, Xi'an 710062, Shaanxi, PR China
| | - Peiyan Yang
- Laboratory of Gene Therapy, Department of Biochemistry, College of Life Sciences, Shaanxi Normal University, 199 South Chang'an Road, Xi'an 710062, Shaanxi, PR China
| | - Qinwen Mao
- Department of Pathology, University of Utah, 2000 Circle of Hope Drive, Salt Lake City, UT 84112, USA
| | - Haibin Xia
- Laboratory of Gene Therapy, Department of Biochemistry, College of Life Sciences, Shaanxi Normal University, 199 South Chang'an Road, Xi'an 710062, Shaanxi, PR China.
| |
Collapse
|
13
|
Zhu G, Yu H, Peng T, Yang K, Xu X, Gu W. Glycolytic enzyme PGK1 promotes M1 macrophage polarization and induces pyroptosis of acute lung injury via regulation of NLRP3. Respir Res 2024; 25:291. [PMID: 39080660 PMCID: PMC11290129 DOI: 10.1186/s12931-024-02926-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Accepted: 07/25/2024] [Indexed: 08/02/2024] Open
Abstract
Acute lung injury (ALI) is characterized by an unregulated inflammatory reaction, often leading to severe morbidity and ultimately death. Excessive inflammation caused by M1 macrophage polarization and pyroptosis has been revealed to have a critical role in ALI. Recent study suggests that glycolytic reprogramming is important in the regulation of macrophage polarization and pyroptosis. However, the particular processes underlying ALI have yet to be identified. In this study, we established a Lipopolysaccharide(LPS)-induced ALI model and demonstrated that blocking glycolysis by using 2-Deoxy-D-glucose(2-DG) significantly downregulated the expression of M1 macrophage markers and pyroptosis-related genes, which was consistent with the in vitro results. Furthermore, our research has revealed that Phosphoglycerate Kinase 1(PGK1), an essential enzyme in the glycolysis pathway, interacts with NOD-, LRR- and pyrin domain-containing protein 3(NLRP3). We discovered that LPS stimulation improves the combination of PGK1 and NLRP3 both in vivo and in vitro. Interestingly, the absence of PGK1 reduces the phosphorylation level of NLRP3. Based on in vitro studies with mice bone marrow-derived macrophages (BMDMs), we further confirmed that siPGK1 plays a protective role by inhibiting macrophage pyroptosis and M1 macrophage polarization. The PGK1 inhibitor NG52 suppresses the occurrence of excessive inflammation in ALI. In general, it is plausible to consider a therapeutic strategy that focuses on modulating the relationship between PGK1 and NLRP3 as a means to mitigate the activation of inflammatory macrophages in ALI.
Collapse
Affiliation(s)
- Guiyin Zhu
- 1Department of Respiratory Medicine, Xinhua hospital, Shanghai Jiao Tong University School of Medicine, 1665 KongJiang road, shanghai, 200092, China
| | - Haiyang Yu
- 1Department of Respiratory Medicine, Xinhua hospital, Shanghai Jiao Tong University School of Medicine, 1665 KongJiang road, shanghai, 200092, China
| | - Tian Peng
- 1Department of Respiratory Medicine, Xinhua hospital, Shanghai Jiao Tong University School of Medicine, 1665 KongJiang road, shanghai, 200092, China
| | - Kun Yang
- 1Department of Respiratory Medicine, Xinhua hospital, Shanghai Jiao Tong University School of Medicine, 1665 KongJiang road, shanghai, 200092, China
| | - Xue Xu
- 1Department of Respiratory Medicine, Xinhua hospital, Shanghai Jiao Tong University School of Medicine, 1665 KongJiang road, shanghai, 200092, China
| | - Wen Gu
- 1Department of Respiratory Medicine, Xinhua hospital, Shanghai Jiao Tong University School of Medicine, 1665 KongJiang road, shanghai, 200092, China.
| |
Collapse
|
14
|
Leishman S, Aljadeed NM, Anand PK. Protocol for a semi-quantitative approach to identify protein S-palmitoylation in cultured cells by acyl biotin exchange assay. STAR Protoc 2024; 5:103054. [PMID: 38704832 PMCID: PMC11089391 DOI: 10.1016/j.xpro.2024.103054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Revised: 03/28/2024] [Accepted: 04/18/2024] [Indexed: 05/07/2024] Open
Abstract
Palmitoylation is a post-translational lipid modification in which palmitic acid is conjugated predominantly to cysteine residues of target proteins, allowing them to tether to cell membranes. Here, we describe a protocol to perform a stepwise acyl biotin exchange assay to identify protein S-palmitoylation. We describe steps for initial blocking of free thiols in protein lysates, subsequent replacement of thioester-linked palmitate groups with a biotin tag for affinity enrichment, and identification of palmitoylated proteins by SDS-PAGE. For complete details on the use and execution of this protocol, please refer to Leishman et al.1.
Collapse
Affiliation(s)
- Stuart Leishman
- Department of Infectious Disease, Imperial College London, London W12 0NN, UK.
| | - Najd M Aljadeed
- Department of Infectious Disease, Imperial College London, London W12 0NN, UK
| | - Paras K Anand
- Department of Infectious Disease, Imperial College London, London W12 0NN, UK.
| |
Collapse
|
15
|
Liu Q, Gu X, Liu X, Gu Y, Zhang H, Yang J, Huang Z. Long-chain fatty acids - The turning point between 'mild' and 'severe' acute pancreatitis. Heliyon 2024; 10:e31296. [PMID: 38828311 PMCID: PMC11140623 DOI: 10.1016/j.heliyon.2024.e31296] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2023] [Revised: 05/14/2024] [Accepted: 05/14/2024] [Indexed: 06/05/2024] Open
Abstract
Acute pancreatitis (AP) is an inflammatory disease characterized by localized pancreatic injury and a systemic inflammatory response. Fatty acids (FAs), produced during the breakdown of triglycerides (TGs) in blood and peripancreatic fat, escalate local pancreatic inflammation to a systemic level by damaging pancreatic acinar cells (PACs) and triggering M1 macrophage polarization. This paper provides a comprehensive analysis of lipases' roles in the onset and progression of AP, as well as the effects of long-chain fatty acids (LCFAs) on the function of pancreatic acinar cells (PACs). Abnormalities in the function of PACs include Ca2+ overload, premature trypsinogen activation, protein kinase C (PKC) expression, endoplasmic reticulum (ER) stress, and mitochondrial and autophagic dysfunction. The study highlights the contribution of long-chain saturated fatty acids (LC-SFAs), especially palmitic acid (PA), to M1 macrophage polarization through the activation of the NLRP3 inflammasome and the NF-κB pathway. Furthermore, we investigated lipid lowering therapy for AP. This review establishes a theoretical foundation for pro-inflammatory mechanisms associated with FAs in AP and facilitating drug development.
Collapse
Affiliation(s)
- Qiang Liu
- Department of Gastroenterology, Affiliated Hangzhou First People's Hospital, Westlake University School of Medicine, Hangzhou 310058, China
- Key Laboratory of Integrated Traditional Chinese and Western Medicine for Biliary and Pancreatic Diseases of Zhejiang Province, Hangzhou 310058, China
- Hangzhou Hospital & Institute of Digestive Diseases, Hangzhou, Zhejiang 310006, China
| | - Xinyi Gu
- The Fourth School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou 310003, China
| | - Xiaodie Liu
- The Fourth School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou 310003, China
| | - Ye Gu
- Department of Gastroenterology, Affiliated Hangzhou First People's Hospital, Westlake University School of Medicine, Hangzhou 310058, China
| | - Hongchen Zhang
- Department of Gastroenterology, Affiliated Hangzhou First People's Hospital, Westlake University School of Medicine, Hangzhou 310058, China
| | - Jianfeng Yang
- Department of Gastroenterology, Affiliated Hangzhou First People's Hospital, Westlake University School of Medicine, Hangzhou 310058, China
- The Fourth School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou 310003, China
- Key Laboratory of Integrated Traditional Chinese and Western Medicine for Biliary and Pancreatic Diseases of Zhejiang Province, Hangzhou 310058, China
- Hangzhou Hospital & Institute of Digestive Diseases, Hangzhou, Zhejiang 310006, China
| | - Zhicheng Huang
- Department of Gastroenterology, Affiliated Hangzhou First People's Hospital, Westlake University School of Medicine, Hangzhou 310058, China
- The Fourth School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou 310003, China
| |
Collapse
|
16
|
Sukhorukov VN, Orekhov AN. Molecular Aspects of Inflammation and Lipid Metabolism in Health and Disease: The Role of the Mitochondria. Int J Mol Sci 2024; 25:6299. [PMID: 38928004 PMCID: PMC11204260 DOI: 10.3390/ijms25126299] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Accepted: 06/04/2024] [Indexed: 06/28/2024] Open
Abstract
Inflammation and lipid metabolism are two deeply interconnected and reciprocally regulated major physiological processes [...].
Collapse
Affiliation(s)
- Vasily N. Sukhorukov
- Petrovsky National Research Centre of Surgery, Moscow 117418, Russia;
- Institute for Atherosclerosis Research, Osennyaya 4-1-207, Moscow 121609, Russia
| | - Alexander N. Orekhov
- Petrovsky National Research Centre of Surgery, Moscow 117418, Russia;
- Institute for Atherosclerosis Research, Osennyaya 4-1-207, Moscow 121609, Russia
| |
Collapse
|
17
|
Yuan Z, Yan R, Fu Z, Wu T, Ren C. Impact of physicochemical properties on biological effects of lipid nanoparticles: Are they completely safe. THE SCIENCE OF THE TOTAL ENVIRONMENT 2024; 927:172240. [PMID: 38582114 DOI: 10.1016/j.scitotenv.2024.172240] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Revised: 04/03/2024] [Accepted: 04/03/2024] [Indexed: 04/08/2024]
Abstract
Lipid nanoparticles (LNPs) are promising materials and human-use approved excipients, with manifold applications in biomedicine. Researchers have tended to focus on improving the pharmacological efficiency and organ targeting of LNPs, while paid relatively less attention to the negative aspects created by their specific physicochemical properties. Here, we discuss the impacts of LNPs' physicochemical properties (size, surface hydrophobicity, surface charge, surface modification and lipid composition) on the adsorption-transportation-distribution-clearance processes and bio-nano interactions. In addition, since there is a lack of review emphasizing on toxicological profiles of LNPs, this review outlined immunogenicity, inflammation, hemolytic toxicity, cytotoxicity and genotoxicity induced by LNPs and the underlying mechanisms, with the aim to understand the properties that underlie the biological effects of these materials. This provides a basic strategy that increased efficacy of medical application with minimized side-effects can be achieved by modulating the physicochemical properties of LNPs. Therefore, addressing the effects of physicochemical properties on toxicity induced by LNPs is critical for understanding their environmental and health risks and will help clear the way for LNPs-based drugs to eventually fulfill their promise as a highly effective therapeutic agents for diverse diseases in clinic.
Collapse
Affiliation(s)
- Ziyi Yuan
- Beijing Key Laboratory of Environmental Toxicology, Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing, China
| | - Ruyu Yan
- Beijing Key Laboratory of Environmental Toxicology, Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing, China
| | - Zuyi Fu
- College of Rehabilitation, Captital Medical University, Beijing, China
| | - Tao Wu
- Beijing Key Laboratory of Enze Biomass Fine Chemicals, College of New Materials and Chemical Engineering, Beijing Institute of Petrochemical Technology, Beijing, China.
| | - Chaoxiu Ren
- Beijing Key Laboratory of Environmental Toxicology, Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing, China.
| |
Collapse
|
18
|
Yang CW, Liu HM, Chang ZY, Liu GH, Chang HH, Huang PY, Lee TY. Puerarin Modulates Hepatic Farnesoid X Receptor and Gut Microbiota in High-Fat Diet-Induced Obese Mice. Int J Mol Sci 2024; 25:5274. [PMID: 38791314 PMCID: PMC11121391 DOI: 10.3390/ijms25105274] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Revised: 05/04/2024] [Accepted: 05/06/2024] [Indexed: 05/26/2024] Open
Abstract
Obesity is associated with alterations in lipid metabolism and gut microbiota dysbiosis. This study investigated the effects of puerarin, a bioactive isoflavone, on lipid metabolism disorders and gut microbiota in high-fat diet (HFD)-induced obese mice. Supplementation with puerarin reduced plasma alanine aminotransferase, liver triglyceride, liver free fatty acid (FFA), and improved gut microbiota dysbiosis in obese mice. Puerarin's beneficial metabolic effects were attenuated when farnesoid X receptor (FXR) was antagonized, suggesting FXR-mediated mechanisms. In hepatocytes, puerarin ameliorated high FFA-induced sterol regulatory element-binding protein (SREBP) 1 signaling, inflammation, and mitochondrial dysfunction in an FXR-dependent manner. In obese mice, puerarin reduced liver damage, regulated hepatic lipogenesis, decreased inflammation, improved mitochondrial function, and modulated mitophagy and ubiquitin-proteasome pathways, but was less effective in FXR knockout mice. Puerarin upregulated hepatic expression of FXR, bile salt export pump (BSEP), and downregulated cytochrome P450 7A1 (CYP7A1) and sodium taurocholate transporter (NTCP), indicating modulation of bile acid synthesis and transport. Puerarin also restored gut microbial diversity, the Firmicutes/Bacteroidetes ratio, and the abundance of Clostridium celatum and Akkermansia muciniphila. This study demonstrates that puerarin effectively ameliorates metabolic disturbances and gut microbiota dysbiosis in obese mice, predominantly through FXR-dependent pathways. These findings underscore puerarin's potential as a therapeutic agent for managing obesity and enhancing gut health, highlighting its dual role in improving metabolic functions and modulating microbial communities.
Collapse
MESH Headings
- Animals
- Isoflavones/pharmacology
- Gastrointestinal Microbiome/drug effects
- Diet, High-Fat/adverse effects
- Receptors, Cytoplasmic and Nuclear/metabolism
- Mice
- Obesity/metabolism
- Obesity/drug therapy
- Liver/metabolism
- Liver/drug effects
- Male
- Dysbiosis
- Mice, Obese
- Mice, Inbred C57BL
- ATP Binding Cassette Transporter, Subfamily B, Member 11/metabolism
- ATP Binding Cassette Transporter, Subfamily B, Member 11/genetics
- Cholesterol 7-alpha-Hydroxylase/metabolism
- Cholesterol 7-alpha-Hydroxylase/genetics
- Mice, Knockout
- Organic Anion Transporters, Sodium-Dependent/metabolism
- Organic Anion Transporters, Sodium-Dependent/genetics
- Symporters/metabolism
- Symporters/genetics
- Lipid Metabolism/drug effects
- Hepatocytes/metabolism
- Hepatocytes/drug effects
- Akkermansia
Collapse
Affiliation(s)
- Ching-Wei Yang
- Graduate Institute of Clinical Medical Sciences, College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan;
- Division of Internal and Pediatric Chinese Medicine, Center for Traditional Chinese Medicine, Chang Gung Memorial Hospital, Linkou 333423, Taiwan
| | - Hsuan-Miao Liu
- Graduate Institute of Traditional Chinese Medicine, School of Chinese Medicine, College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan;
| | - Zi-Yu Chang
- Department of Traditional Chinese Medicine, Chang Gung Memorial Hospital, Keelung 20401, Taiwan;
| | - Geng-Hao Liu
- School of Traditional Chinese Medicine, College of Medicine, Chang Gung University, Taoyuan 333323, Taiwan;
- Division of Acupuncture and Moxibustion, Center for Traditional Chinese Medicine, Chang Gung Memorial Hospital, Taoyuan 333423, Taiwan
- Sleep Center, Chang Gung Memorial Hospital, Taoyuan 333008, Taiwan
| | - Hen-Hong Chang
- Graduate Institute of Integrated Medicine, China Medical University, Taichung 40402, Taiwan;
| | - Po-Yu Huang
- Department of Chinese Medicine, Linsen Chinese Medicine and Kunming Branch, Taipei City Hospital, Taipei 10844, Taiwan
| | - Tzung-Yan Lee
- Graduate Institute of Traditional Chinese Medicine, School of Chinese Medicine, College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan;
- School of Traditional Chinese Medicine, College of Medicine, Chang Gung University, Taoyuan 333323, Taiwan;
| |
Collapse
|
19
|
Huang H, Sharoar MG, Pathoulas J, Fan L, He W, Xiang R, Yan R. Accumulation of neutral lipids in dystrophic neurites surrounding amyloid plaques in Alzheimer's disease. Biochim Biophys Acta Mol Basis Dis 2024; 1870:167086. [PMID: 38378084 PMCID: PMC10999334 DOI: 10.1016/j.bbadis.2024.167086] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2023] [Revised: 02/06/2024] [Accepted: 02/15/2024] [Indexed: 02/22/2024]
Abstract
Alzheimer's disease (AD) is characterized by the formation β-amyloid (Aβ) deposited neuritic plaques. Recent evidence suggests that abnormal lipid metabolism and accumulation could serve as biomarkers for neurodegenerative diseases, including AD. Tubular endoplasmic reticulum protein, reticulon 3 (RTN3), plays a crucial role in the development of neuritic plaque and lipid metabolism in AD brains. In present study, we sought to investigate a potential association between neutral lipid accumulation and AD pathology. BODIPY 500/510 dye was used to label neutral lipid surrounding Aβ plaques in APPNL-G-F mouse and AD postmortem brains samples. Immunofluorescent images were captured using confocal microscope and co-localization between lipid metabolism proteins and neutral lipids were evaluated. Lipid accumulation in Aβ plaque surrounding dystrophic neurites (DNs) was observed in the cortical region of AD mouse models and human AD brain samples. The neutral lipid staining was not co-localized with IBA1-labeled microglia or GFAP-labeled astrocytes, but it was co-labeled with VAMP2 and neurofilament. We further showed that neutral lipids were accumulated in RTN3 immunoreactive DNs. Both the neutral lipids accumulation and RIDNs formation showed age-dependent patterns in surrounding amyloid plaques. Mechanistic studies revealed that RTN3 likely contributes to the enrichment of neutral lipids near plaques by interacting with heat shock cognate protein 70 (HSC70) and diminishing its function in chaperone-mediated lipophagy. Our study provides immunohistochemical evidence of neutral lipids being enriched in DNs near amyloid plaques. Our findings shed light on RTN3-mediaed lipid accumulation in AD neuropathology and provide fresh insights into the role of RTN3 in neurodegenerative diseases.
Collapse
Affiliation(s)
- Hao Huang
- Department of Nephrology, Xiangya Hospital and National Clinical Research Center for Geriatric Disorders, Central South University, Changsha, China; Department of Neuroscience, University of Connecticut Health, Farmington, CT, USA; Department of Cell Biology, School of Life Sciences, Central South University, Changsha, China; Hunan Key Laboratory of Organ Fibrosis, Central South University, Changsha, China.
| | - Md Golam Sharoar
- Department of Neuroscience, University of Connecticut Health, Farmington, CT, USA; Alzheimer's Disease Research Program, Corewell Health Research Institute, Oakland University William Beaumont School of Medicine, Corewell Health East, Royal Oak, MI 48073, USA
| | - Joseph Pathoulas
- Department of Neuroscience, University of Connecticut Health, Farmington, CT, USA
| | - Liangliang Fan
- Department of Cell Biology, School of Life Sciences, Central South University, Changsha, China
| | - Wanxia He
- Department of Neuroscience, University of Connecticut Health, Farmington, CT, USA
| | - Rong Xiang
- Department of Cell Biology, School of Life Sciences, Central South University, Changsha, China; Hunan Key Laboratory of Organ Fibrosis, Central South University, Changsha, China.
| | - Riqiang Yan
- Department of Neuroscience, University of Connecticut Health, Farmington, CT, USA.
| |
Collapse
|
20
|
Schwärzler J, Grabherr F, Grander C, Adolph TE, Tilg H. The pathophysiology of MASLD: an immunometabolic perspective. Expert Rev Clin Immunol 2024; 20:375-386. [PMID: 38149354 DOI: 10.1080/1744666x.2023.2294046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Accepted: 12/08/2023] [Indexed: 12/28/2023]
Abstract
INTRODUCTION Metabolic-associated liver diseases have emerged pandemically across the globe and are clinically related to metabolic disorders such as obesity and type 2 diabetes. The new nomenclature and definition (i.e. metabolic dysfunction-associated steatotic liver disease - MASLD; metabolic dysfunction-associated steatohepatitis - MASH) reflect the nature of these complex systemic disorders, which are characterized by inflammation, gut dysbiosis and metabolic dysregulation. In this review, we summarize recent advantages in understanding the pathophysiology of MASLD, which we parallel to emerging therapeutic concepts. AREAS COVERED We summarize the pathophysiologic concepts of MASLD and its transition to MASH and subsequent advanced sequelae of diseases. Furthermore, we highlight how dietary constituents, microbes and associated metabolites, metabolic perturbations, and immune dysregulation fuel lipotoxicity, hepatic inflammation, liver injury, insulin resistance, and systemic inflammation. Deciphering the intricate pathophysiologic processes that contribute to the development and progression of MASLD is essential to develop targeted therapeutic approaches to combat this escalating burden for health-care systems. EXPERT OPINION The rapidly increasing prevalence of metabolic dysfunction-associated steatotic liver disease challenges health-care systems worldwide. Understanding pathophysiologic traits is crucial to improve the prevention and treatment of this disorder and to slow progression into advanced sequelae such as cirrhosis and hepatocellular carcinoma.
Collapse
Affiliation(s)
- Julian Schwärzler
- Department of Internal Medicine I, Gastroenterology, Hepatology, Endocrinology & Metabolism, Medical University of Innsbruck, Innsbruck, Austria
| | - Felix Grabherr
- Department of Internal Medicine I, Gastroenterology, Hepatology, Endocrinology & Metabolism, Medical University of Innsbruck, Innsbruck, Austria
| | - Christoph Grander
- Department of Internal Medicine I, Gastroenterology, Hepatology, Endocrinology & Metabolism, Medical University of Innsbruck, Innsbruck, Austria
| | - Timon E Adolph
- Department of Internal Medicine I, Gastroenterology, Hepatology, Endocrinology & Metabolism, Medical University of Innsbruck, Innsbruck, Austria
| | - Herbert Tilg
- Department of Internal Medicine I, Gastroenterology, Hepatology, Endocrinology & Metabolism, Medical University of Innsbruck, Innsbruck, Austria
| |
Collapse
|
21
|
Wu Y, Zhou J, Zuo X, Kuang Y, Sun L, Zhang X. Yanggan Jiangmei Formula alleviates hepatic inflammation and lipid accumulation in non-alcoholic steatohepatitis by inhibiting the NF-κB/NLRP3 signaling pathway. Chin J Nat Med 2024; 22:224-234. [PMID: 38553190 DOI: 10.1016/s1875-5364(24)60595-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Indexed: 04/02/2024]
Abstract
The role of NF-κB and the NLRP3 inflammasome in the chronic inflammatory microenvironment of non-alcoholic steatohepatitis (NASH) has been posited as crucial. The Yanggan Jiangmei Formula (YGJMF) has shown promise in ameliorating hepatic steatosis in NASH patients, yet its pharmacological mechanisms remain largely unexplored. This study was conducted to investigate the efficacy of YGJMF in NASH and to elucidate its pharmacological underpinnings. To simulate NASH both in vivo and in vitro, high-fat-diet (HFD) rats and HepG2 cells stimulated with free fatty acids (FFAs) were utilized. The severity of liver injury and lipid deposition was assessed using serum indicators, histopathological staining, micro-magnetic resonance imaging (MRI), and the liver-to-muscle signal intensity ratio (SIRL/M). Furthermore, a combination of enzyme-linked immunosorbent assay (ELISA), immunohistochemistry (IHC), immunofluorescence, real-time quantitative polymerase chain reaction (RT-qPCR), and Western blotting analyses was employed to investigate the NF-κB/NLRP3 signaling pathway and associated cytokine levels. The results from liver pathology, MRI assessments, and biochemical tests in rat models demonstrated YGJMF's significant effectiveness in reducing liver damage and lipid accumulation. Additionally, YGJMF markedly reduced hepatocyte inflammation by downregulating inflammatory cytokines in both liver tissue and serum. Furthermore, YGJMF was found to disrupt NF-κB activation, consequently inhibiting the assembly of the NLRP3 inflammasome in both the in vitro and in vivo models. The preliminary findings of this study suggest that YGJMF may alleviate hepatic steatosis and inhibit the NF-κB/NLRP3 signaling pathway, thereby exerting anti-inflammatory effects in NASH.
Collapse
Affiliation(s)
- Yuanyuan Wu
- The First Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing 210023, China; The Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Nanjing 210029, China
| | - Jingwen Zhou
- The First Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Xinchen Zuo
- The First Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing 210023, China; The Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Nanjing 210029, China
| | - Yufeng Kuang
- The First Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing 210023, China; The Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Nanjing 210029, China
| | - Lixia Sun
- The First Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing 210023, China; The Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Nanjing 210029, China.
| | - Xiaolong Zhang
- The Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Nanjing 210029, China.
| |
Collapse
|
22
|
Tang B, Zheng X, Luo Q, Li X, Yang Y, Bi Y, Chen Y, Han L, Chen H, Lu C. Network pharmacology and gut microbiota insights: unraveling Shenling Baizhu powder's role in psoriasis treatment. Front Pharmacol 2024; 15:1362161. [PMID: 38425649 PMCID: PMC10904012 DOI: 10.3389/fphar.2024.1362161] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2023] [Accepted: 01/30/2024] [Indexed: 03/02/2024] Open
Abstract
Background: Psoriasis, a chronic skin condition characterized by systemic inflammation and altered gut microbiota, has been a target of Traditional Chinese Medicine (TCM) for centuries. Shenling Baizhu Powder (SLBZP), a TCM formulation, holds promise for treating inflammatory diseases, but its specific role in psoriasis and impact on gut microbiota is not fully understood. Objective: This study aims to elucidate the mechanism of SLBZP in treating psoriasis, integrating component analysis, network pharmacology, and experimental validation in mice models. Methods: We commenced with a detailed component analysis of SLBZP using liquid chromatograph and mass spectrometer (LC-MS). Network pharmacology analysis was used to predict the potential action targets and pathways of SLBZP in psoriasis. An in vivo experiment was conducted with psoriasis mice models, treated with SLBZP. Therapeutic effects were assessed via symptomatology, histopathology, and immunohistochemical analysis. Gut microbiota composition was analyzed using 16S rRNA gene sequencing. Results: A total of 42 main components and quality markers were identified, primarily from licorice and ginseng, including flavonoids, saponins and other markers. PPI topology analysis showed that TNF, IL-6, IL-1β, TP53 and JUN were the core DEPs. 168 signaling pathways including lipid and atherosclerosis, AGE-RAGE signaling pathway, IL-17 signaling pathway and Th17 cell differentiation were enriched by KEGG. SLBZP demonstrated significant therapeutic effects on psoriasis in mice, with alterations in skin pathology and biomarkers. Additionally, notable changes in gut microbiota composition were observed post-treatment, indicating a possible gut-skin axis involvement. Conclusion: This research has pinpointed lipid metabolism as a key pathway in the treatment of psoriasis with SLBZP. It explores how SLBZP's modulation of gut microbiota and lipid metabolism can alleviate psoriasis, suggesting that balancing gut microbiota may reduce inflammation mediators and offer therapeutic benefits. This underscores lipid metabolism modulation as a potential new strategy in psoriasis treatment.
Collapse
Affiliation(s)
- Bin Tang
- The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, China
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, Guangdong Provincial Hospital of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
- Guangdong Provincial Clinical Medicine Research Center for Chinese Medicine Dermatology, Guangzhou, China
- Guangdong-Hong Kong-Macau Joint Lab on Chinese Medicine and Immune Disease Research, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Xuwei Zheng
- The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Qianqian Luo
- The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Xiong Li
- The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yujie Yang
- The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yang Bi
- The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yonggen Chen
- The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Ling Han
- The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, China
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, Guangdong Provincial Hospital of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
- Guangdong Provincial Clinical Medicine Research Center for Chinese Medicine Dermatology, Guangzhou, China
- Guangdong-Hong Kong-Macau Joint Lab on Chinese Medicine and Immune Disease Research, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Haiming Chen
- The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, China
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, Guangdong Provincial Hospital of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
- Guangdong Provincial Clinical Medicine Research Center for Chinese Medicine Dermatology, Guangzhou, China
- Guangdong-Hong Kong-Macau Joint Lab on Chinese Medicine and Immune Disease Research, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Chuanjian Lu
- The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, China
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, Guangdong Provincial Hospital of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
- Guangdong Provincial Clinical Medicine Research Center for Chinese Medicine Dermatology, Guangzhou, China
- Guangdong-Hong Kong-Macau Joint Lab on Chinese Medicine and Immune Disease Research, Guangzhou University of Chinese Medicine, Guangzhou, China
| |
Collapse
|
23
|
Jiao R, Jiang W, Xu K, Luo Q, Wang L, Zhao C. Lipid metabolism analysis in esophageal cancer and associated drug discovery. J Pharm Anal 2024; 14:1-15. [PMID: 38352954 PMCID: PMC10859535 DOI: 10.1016/j.jpha.2023.08.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Revised: 07/27/2023] [Accepted: 08/29/2023] [Indexed: 02/16/2024] Open
Abstract
Esophageal cancer is an upper gastrointestinal malignancy with a bleak prognosis. It is still being explored in depth due to its complex molecular mechanisms of occurrence and development. Lipids play a crucial role in cells by participating in energy supply, biofilm formation, and signal transduction processes, and lipid metabolic reprogramming also constitutes a significant characteristic of malignant tumors. More and more studies have found esophageal cancer has obvious lipid metabolism abnormalities throughout its beginning, progress, and treatment resistance. The inhibition of tumor growth and the enhancement of antitumor therapy efficacy can be achieved through the regulation of lipid metabolism. Therefore, we reviewed and analyzed the research results and latest findings for lipid metabolism and associated analysis techniques in esophageal cancer, and comprehensively proved the value of lipid metabolic reprogramming in the evolution and treatment resistance of esophageal cancer, as well as its significance in exploring potential therapeutic targets and biomarkers.
Collapse
Affiliation(s)
- Ruidi Jiao
- Bionic Sensing and Intelligence Center, Institute of Biomedical and Health Engineering, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong, 518000, China
- Department of Radiation Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital & Shenzhen Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shenzhen, Guangdong, 518116, China
- School of Medicine, Southern University of Science and Technology, Shenzhen, Guangdong, 518000, China
| | - Wei Jiang
- Department of Radiation Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital & Shenzhen Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shenzhen, Guangdong, 518116, China
| | - Kunpeng Xu
- Department of Radiation Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital & Shenzhen Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shenzhen, Guangdong, 518116, China
| | - Qian Luo
- Bionic Sensing and Intelligence Center, Institute of Biomedical and Health Engineering, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong, 518000, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Luhua Wang
- Department of Radiation Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital & Shenzhen Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shenzhen, Guangdong, 518116, China
- School of Medicine, Southern University of Science and Technology, Shenzhen, Guangdong, 518000, China
| | - Chao Zhao
- Bionic Sensing and Intelligence Center, Institute of Biomedical and Health Engineering, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong, 518000, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
- Shenzhen Key Laboratory of Precision Diagnosis and Treatment of Depression, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong, 518000, China
| |
Collapse
|
24
|
Wang Y, Xie H, Huang X, Chen K, Zhu Y, Yao G. Retrospective analysis and preliminary laboratory validation of treatment efficacy and blood lipid levels in patients with rheumatoid arthritis. Clin Rheumatol 2023; 42:3213-3223. [PMID: 37488372 DOI: 10.1007/s10067-023-06683-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2023] [Revised: 06/16/2023] [Accepted: 06/19/2023] [Indexed: 07/26/2023]
Abstract
OBJECTIVE Rheumatoid arthritis is a systemic disease characterized by progressive chronic inflammation resulting in destruction of synovial joints. In addition to joint involvement, abnormal blood lipid indexes have also been found in RA patients. The correlation between various blood lipid indexes and the treatment effects were assessed in patients with rheumatoid arthritis, for the purposes to find a better medication strategy for RA. METHODS One hundred nineteen rheumatoid arthritis patients were recruited and divided into two groups, 45 patients with significant drug treatment effect and 45 patients with insignificant drug treatment effect through the nearest neighbor matching method in propensity score. The correlation between various blood lipid indexes and drug treatment effect of rheumatoid arthritis patients was analyzed. A mouse model of rheumatoid arthritis was constructed in the laboratory; methotrexate was treated as a positive drug. We observe and record the onset of rheumatoid arthritis in mice, as well as the proportion of immune cells, the expression of inflammatory factors, and the changes in blood lipid profiles was done. RESULTS The levels of total cholesterol (TC), triglyceride (TG), high density lipoprotein (HDL-C), low density lipoprotein (LDL-C), and erythrocyte sedimentation rate (ESR) in rheumatoid arthritis patients were significantly different between the two groups (P < 0.05). There was no significant difference in other indexes between the two groups (P > 0.05). Methotrexate had a good therapeutic effect on CIA model mice, and the levels of TC and HDL-C in the treatment group were higher than those in the model group. CONCLUSION There is a high correlation between the levels of TC and HDL-C in rheumatoid arthritis patients and the effect of drug treatment. In the clinical treatment of rheumatoid arthritis, we should focus on improving the blood lipid indexes such as TC and HDL-C, and explore more targeted individualized administration, so as to achieve better and faster treatment effect in patients with rheumatoid arthritis. Key Points • In this research, we found that the TC and HDL-C level in RA patients' blood is highly related with the therapeutic effect, and a lower level of TC and HDL-C is better for therapeutic effect of RA.
Collapse
Affiliation(s)
- Yuchun Wang
- State Key Laboratory of Quality Research in Chinese Medicine and School of Pharmacy, Macau University of Science and Technology, Macau, 999078, China
- Department of Rheumatology and Immunology, The Affiliated Drum Tower Hospital of Nanjing University, Nanjing, 210093, China
| | - Han Xie
- State Key Laboratory of Quality Research in Chinese Medicine and School of Pharmacy, Macau University of Science and Technology, Macau, 999078, China
- Department of Rheumatology and Immunology, The Affiliated Drum Tower Hospital of Nanjing University, Nanjing, 210093, China
| | - Xuan Huang
- Department of Rheumatology and Immunology, The Affiliated Drum Tower Hospital of Nanjing University, Nanjing, 210093, China
| | - Keyuan Chen
- State Key Laboratory of Quality Research in Chinese Medicine and School of Pharmacy, Macau University of Science and Technology, Macau, 999078, China
| | - Yizhun Zhu
- State Key Laboratory of Quality Research in Chinese Medicine and School of Pharmacy, Macau University of Science and Technology, Macau, 999078, China.
| | - Genhong Yao
- Department of Rheumatology and Immunology, The Affiliated Drum Tower Hospital of Nanjing University, Nanjing, 210093, China
| |
Collapse
|
25
|
Guo Z, Yiu N, Hu Z, Zhou W, Long X, Yang M, Liao J, Zhang G, Lu Q, Zhao M. Alterations of fecal microbiome and metabolome in pemphigus patients. J Autoimmun 2023; 141:103108. [PMID: 37714737 DOI: 10.1016/j.jaut.2023.103108] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Revised: 07/12/2023] [Accepted: 08/24/2023] [Indexed: 09/17/2023]
Abstract
The role of gut microbiome and metabolic substances in the development of autoimmune diseases has gradually been revealed. However, the relevant gut features in pemphigus have not been well clarified. We collected stool samples from pemphigus patients and healthy controls (HCs). Metagenomic sequencing and liquid chromatography-mass spectrometry (LC/MS) metabolome sequencing were performed to analyze the compositional and metabolic alternations of the gut microbiome in pemphigus patients and HCs. We observed the reduced richness and diversity and greater heterogeneity in pemphigus patients, which was characterized by a significant decrease in Firmicutes and an increase in Proteobacteria. At the species level, Intestinal pathogenic bacteria such as Escherichia coli and Bacteroides fragilis were significantly enriched, while anti-inflammatory bacteria and butyric acid-producing bacteria were significantly reduced, which were related to clinical indicators (Dsg1/3 and PDAI). 4 species were selected by the machine learning algorithm to better distinguish pemphigus patients from healthy people. Metabolomic analysis showed that the composition of pemphigus patients was different from that of HCs. PE (18:3 (6Z,9Z, 12Z)/14:1 (9Z)) was the main metabolic substance in pemphigus and involved in a variety of metabolic pathways. While Retinol, flavonoid compounds and various amino acids decreased significantly compared with HCs. Furthermore, we found that differences in the levels of these metabolites correlated with changes in the abundance of specific species. Our study provides a comprehensive picture of gut microbiota and metabolites in pemphigus patients and suggests a potential mechanism of the aberrant gut microbiota and metabolites in the pathogenesis of pemphigus.
Collapse
Affiliation(s)
- Ziyu Guo
- Department of Dermatology, Hunan Key Laboratory of Medical Epigenomics, The Second Xiangya Hospital of Central, South University, Changsha, Hunan, China; Clinical Medical Research Center of Major Skin Diseases and Skin Health of Hunan Province, Changsha, China
| | - Nam Yiu
- Department of Dermatology, Hunan Key Laboratory of Medical Epigenomics, The Second Xiangya Hospital of Central, South University, Changsha, Hunan, China; Clinical Medical Research Center of Major Skin Diseases and Skin Health of Hunan Province, Changsha, China
| | - Zhi Hu
- Department of Dermatology, Hunan Key Laboratory of Medical Epigenomics, The Second Xiangya Hospital of Central, South University, Changsha, Hunan, China; Clinical Medical Research Center of Major Skin Diseases and Skin Health of Hunan Province, Changsha, China
| | - Wenyu Zhou
- Department of Dermatology, Hunan Key Laboratory of Medical Epigenomics, The Second Xiangya Hospital of Central, South University, Changsha, Hunan, China; Clinical Medical Research Center of Major Skin Diseases and Skin Health of Hunan Province, Changsha, China
| | - Xuan Long
- Department of Dermatology, Hunan Key Laboratory of Medical Epigenomics, The Second Xiangya Hospital of Central, South University, Changsha, Hunan, China; Clinical Medical Research Center of Major Skin Diseases and Skin Health of Hunan Province, Changsha, China
| | - Miao Yang
- Department of Dermatology, Hunan Key Laboratory of Medical Epigenomics, The Second Xiangya Hospital of Central, South University, Changsha, Hunan, China; Clinical Medical Research Center of Major Skin Diseases and Skin Health of Hunan Province, Changsha, China
| | - Jieyue Liao
- Department of Dermatology, Hunan Key Laboratory of Medical Epigenomics, The Second Xiangya Hospital of Central, South University, Changsha, Hunan, China; Clinical Medical Research Center of Major Skin Diseases and Skin Health of Hunan Province, Changsha, China
| | - Guiying Zhang
- Department of Dermatology, Hunan Key Laboratory of Medical Epigenomics, The Second Xiangya Hospital of Central, South University, Changsha, Hunan, China; Clinical Medical Research Center of Major Skin Diseases and Skin Health of Hunan Province, Changsha, China.
| | - Qianjin Lu
- Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing 210042, China; Key Laboratory of Basic and Translational Research on Immune-Mediated Skin Diseases, Chinese Academy of Medical Sciences, Nanjing, China.
| | - Ming Zhao
- Department of Dermatology, Hunan Key Laboratory of Medical Epigenomics, The Second Xiangya Hospital of Central, South University, Changsha, Hunan, China; Clinical Medical Research Center of Major Skin Diseases and Skin Health of Hunan Province, Changsha, China; Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing 210042, China; Key Laboratory of Basic and Translational Research on Immune-Mediated Skin Diseases, Chinese Academy of Medical Sciences, Nanjing, China.
| |
Collapse
|
26
|
Qiu J, Chen L, Zhang L, Xu F, Zhang C, Ren G, Chang K, He G, Du Z, Le Y, Yu Z, Li S, Liu Q, Dou X. Xie Zhuo Tiao Zhi formula modulates intestinal microbiota and liver purine metabolism to suppress hepatic steatosis and pyroptosis in NAFLD therapy. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2023; 121:155111. [PMID: 37804819 DOI: 10.1016/j.phymed.2023.155111] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Revised: 09/05/2023] [Accepted: 09/19/2023] [Indexed: 10/09/2023]
Abstract
BACKGROUND Current evidence indicates a rising global prevalence of Non-Alcoholic Fatty Liver Disease (NAFLD), which is closely associated to conditions such as obesity, dyslipidemia, insulin resistance, and metabolic syndrome. The relationship between the gut microbiome and metabolites in NAFLD is gaining attention understanding the pathogenesis and progression of dysregulated lipid metabolism and inflammation. The Xie Zhuo Tiao Zhi (XZTZ) decoction has been employed in clinical practice for alleviating hyperlipidemia and symptoms related to metabolic disorders. However, the pharmacological mechanisms underlying the effects of XZTZ remain to be elucidated. PURPOSE The objective of this study was to examine the pharmacological mechanisms underlying the hypolipidemic and anti-inflammatory effects of XZTZ decoction in a mouse model of NAFLD, as well as the effects of supplementing exogenous metabolites on PO induced cell damage and lipid accumulation in cultured hepatocytes. METHODS A high-fat diet (HFD) mouse model was established to examine the effects of XZTZ through oral gavage. The general condition of mice and the protective effect of XZTZ on liver injury were evaluated using histological and biochemical methods. Hematoxylin and eosin staining (H&E) staining and oil red O staining were performed to assess inflammatory and lipid accumulation detection, and cytokine levels were quantitatively analyzed. Additionally, the study included full-length 16S rRNA sequencing, liver transcriptome analysis, and non-targeted metabolomics analysis to investigate the relationship among intestinal microbiome, liver metabolic function, and XZTZ decoction. RESULTS XZTZ had a significant impact on the microbial community structure in NAFLD mice. Notably, the abundance of Ileibacterium valens, which was significantly enriched by XZTZ, exhibited a negative correlation with liver injury biomarkers such as, alanine transaminase (ALT) and aspartate transaminase (AST) activity. Moreover, treatment with XZTZ led to a significant enrichment of the purine metabolism pathway in liver tissue metabolites, with inosine, a purine metabolite, showing a significant positive correlation with the abundance of I. valens. XZTZ and inosine also significantly enhanced fatty acid β-oxidation, which led to a reduction in the expression of pro-inflammatory cytokines and the inhibition of liver pyroptosis. These effects contributed to the mitigation of liver injury and hepatocyte damage, both in vivo and vitro. Furthermore, the utilization of HPLC fingerprints and UPLC-Q-TOF-MS elucidated the principal constituents within the XZTZ decoction, including naringin, neohesperidin, atractylenolide III, 23-o-Acetylalisol B, pachymic acid, and ursolic acid which are likely responsible for its therapeutic efficacy. Further investigations are imperative to fully uncover and validate the pharmacodynamic mechanisms underlying these observations. CONCLUSION The administration of XZTZ decoction demonstrates a protective effect on the livers of NAFLD mice by inhibiting lipid accumulation and reducing hepatocyte inflammatory damage. This protective effect is mediated by the upregulation of I.valens abundance in the intestine, highlighting the importance of the gut-liver axis. Furthermore, the presesnce of inosine, adenosine, and their derivatives are important in promoting the protective effects of XZTZ. Furthermore, the in vitro approaching, we provide hitherto undocumented evidence indicating that the inosine significantly improves lipid accumulation, inflammatory damage, and pyroptosis in AML12 cells incubated with free fatty acids.
Collapse
Affiliation(s)
- Jiannan Qiu
- School of Life Science, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, PR China
| | - Lin Chen
- School of Life Science, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, PR China
| | - Ling Zhang
- School of Life Science, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, PR China
| | - Fangying Xu
- School of Life Science, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, PR China
| | - Congcong Zhang
- School of Life Science, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, PR China
| | - Guilin Ren
- School of Life Science, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, PR China
| | - Kaixin Chang
- School of Life Science, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, PR China
| | - Guonong He
- Ningbo Traditional Chinese Medicine Hospital, Ningbo, Zhejiang, PR China
| | - Zhongyan Du
- School of Basic Medicine, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, PR China
| | - Yifei Le
- School of Life Science, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, PR China
| | - Zhiling Yu
- Consun Chinese Medicines Research Centre for Renal Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China
| | - Songtao Li
- School of Public Health, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, PR China
| | - Qingsheng Liu
- Hangzhou Hospital of Traditional Chinese Medicine Affiliated to Zhejiang University of Traditional Chinese Medicine, Hangzhou, Zhejiang, PR China.
| | - Xiaobing Dou
- School of Life Science, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, PR China.
| |
Collapse
|
27
|
Xu H, Zhong X, Wang T, Wu S, Guan H, Wang D. (-)-Epigallocatechin-3-Gallate Reduces Perfluorodecanoic Acid-Exacerbated Adiposity and Hepatic Lipid Accumulation in High-Fat Diet-Fed Male C57BL/6J Mice. Molecules 2023; 28:7832. [PMID: 38067561 PMCID: PMC10708200 DOI: 10.3390/molecules28237832] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Revised: 11/20/2023] [Accepted: 11/20/2023] [Indexed: 12/18/2023] Open
Abstract
Perfluorodecanoic acid (PFDA), an enduring and harmful organic pollutant, is widely employed in diverse food-related sectors. Our previous studies have provided evidence that PFDA has the potential to facilitate obesity and hepatic fat accumulation induced by high-fat diet (HFD) intake. Epigallocatechin-3-gallate (EGCG), a polyphenol found in green tea, has been suggested to possess potential preventive effects against metabolic abnormalities and fatty liver. The purpose of this research was to investigate the effects of EGCG on PFDA-exacerbated adiposity and hepatic lipid accumulation in HFD-fed mice. The results showed that EGCG reduced body weight gain; tissue and organ weights; blood glucose, serum insulin, HOMA-IR, leptin, and lipid parameters; serum inflammatory cytokines (IL-1β, IL-18, IL-6, and TNF-α); and hepatic lipid accumulation in PFDA-exposed mice fed an HFD. Further work showed that EGCG improved liver function and glucose homeostasis in mice fed an HFD and co-exposed to PFDA. The elevated hepatic mRNA levels of SREBP-1 and associated lipogenic genes, NLRP3, and caspase-1 in PFDA-exposed mice fed an HFD were significantly decreased by EGCG. Our work provides evidence for the potential anti-obesity effect of EGCG on co-exposure to HFD and PFDA and may call for further research on the bioactivity of EGCG to attenuate the endocrine disruption effects of long-term exposure to pollutants.
Collapse
Affiliation(s)
- Hong Xu
- School of Grain Science and Technology, Jiangsu University of Science and Technology, Zhenjiang 212100, China; (H.X.); (X.Z.)
| | - Xu Zhong
- School of Grain Science and Technology, Jiangsu University of Science and Technology, Zhenjiang 212100, China; (H.X.); (X.Z.)
| | - Taotao Wang
- Department of Clinical Nutrition, Affiliated Hospital of Jiangsu University, Zhenjiang 212000, China;
| | - Shanshan Wu
- College of Agriculture & Biotechnology, Zhejiang University, Hangzhou 310058, China;
| | - Huanan Guan
- School of Grain Science and Technology, Jiangsu University of Science and Technology, Zhenjiang 212100, China; (H.X.); (X.Z.)
| | - Dongxu Wang
- School of Grain Science and Technology, Jiangsu University of Science and Technology, Zhenjiang 212100, China; (H.X.); (X.Z.)
| |
Collapse
|
28
|
Wang Y, Cai L, Li H, Chen H, Yang T, Tan Y, Guo Z, Wang X. Overcoming Cancer Resistance to Platinum Drugs by Inhibiting Cholesterol Metabolism. Angew Chem Int Ed Engl 2023; 62:e202309043. [PMID: 37612842 DOI: 10.1002/anie.202309043] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Revised: 08/18/2023] [Accepted: 08/21/2023] [Indexed: 08/25/2023]
Abstract
Drug resistance is a serious challenge for platinum anticancer drugs. Platinum complexes may get over the drug resistance via a distinct mechanism of action. Cholesterol is a key factor contributing to the drug resistance. Inhibiting cellular cholesterol synthesis and uptake provides an alternative strategy for cancer treatment. Platinum(IV) complexes FP and DFP with fenofibric acid as axial ligand(s) were designed to combat the drug resistance through regulating cholesterol metabolism besides damaging DNA. In addition to producing reactive oxygen species and active platinum(II) species to damage DNA, FP and DFP inhibited cellular cholesterol accumulation, promoted cholesterol efflux, upregulated peroxisome proliferator-activated receptor alpha (PPARα), induced caspase-1 activation and gasdermin D (GSDMD) cleavage, thus leading to both apoptosis and pyroptosis in cancer cells. The reduction of cholesterol significantly relieved the drug resistance of cancer cells. The double-acting mechanism gave the complexes strong anticancer activity in vitro and in vivo, particularly against cisplatin-resistant cancer cells.
Collapse
Affiliation(s)
- Ying Wang
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, 210023, P. R. China
| | - Linxiang Cai
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, 210023, P. R. China
| | - Hui Li
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, 210023, P. R. China
| | - Hanhua Chen
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, 210023, P. R. China
| | - Tao Yang
- State Key Laboratory of Coordination Chemistry, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing, 210023, P. R. China
| | - Yehong Tan
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, 210023, P. R. China
| | - Zijian Guo
- State Key Laboratory of Coordination Chemistry, School of Chemistry and Chemical Engineering, Nanjing University, Nanjing, 210023, P. R. China
| | - Xiaoyong Wang
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, 210023, P. R. China
| |
Collapse
|
29
|
Hu S, Li W, Cai Z, Tang C, Li B, Liu S, Li Y. Research progress on chitin/chitosan-based emulsion delivery systems and their application in lipid digestion regulation. Crit Rev Food Sci Nutr 2023; 64:13275-13297. [PMID: 37811646 DOI: 10.1080/10408398.2023.2264392] [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: 10/10/2023]
Abstract
Excessive lipid intake is linked to an elevated risk of health problems. However, reducing lipid contents may influence food structure and flavor. Some alternatives are needed to control the lipid absorption. Emulsions are common carriers for lipids, which can control the hydrolysis and absorption of lipids. Chitin (Ch) and chitosan (CS) are natural polysaccharides with good biodegradability, biocompatibility, and unique cationic properties. They have been reported to be able to delay lipolysis, which can be regarded as one of the most promising agents that regulates lipid digestion (LiD). The application of Ch/CS and their derivatives in emulsions are summarized in this review with a focus on their performances and mechanisms for LiD regulation, aiming to provide theoretical guidance for the development of novel Ch/CS emulsions, and the regulation of LiD. A reasonable design of emulsion interface can provide its resistance to the external environment and then control LiD. The properties of emulsion interface are the key factors affecting LiD. Therefore, systematic study on the relationship between Ch/CS-based emulsion structure and LiD can not only instruct the reasonable design of emulsion interface to accurately regulate LiD, but also provide scientific guidelines for applying Ch/CS in functional food, medicine and other fields.
Collapse
Affiliation(s)
- Shanshan Hu
- College of Food Science and Technology, Huazhong Agricultural University, Wuhan, China
| | - Wenbo Li
- College of Food Science and Technology, Huazhong Agricultural University, Wuhan, China
| | - Zhi Cai
- School of Life Science and Technology, Wuhan Polytechnic University, Wuhan, China
| | - Cuie Tang
- College of Food Science and Technology, Huazhong Agricultural University, Wuhan, China
| | - Bin Li
- College of Food Science and Technology, Huazhong Agricultural University, Wuhan, China
- Key Laboratory of Environment Correlative Dietology, Huazhong Agricultural University, Ministry of Education, Wuhan, China
| | - Shilin Liu
- College of Food Science and Technology, Huazhong Agricultural University, Wuhan, China
- Key Laboratory of Environment Correlative Dietology, Huazhong Agricultural University, Ministry of Education, Wuhan, China
| | - Yan Li
- College of Food Science and Technology, Huazhong Agricultural University, Wuhan, China
- Key Laboratory of Environment Correlative Dietology, Huazhong Agricultural University, Ministry of Education, Wuhan, China
| |
Collapse
|
30
|
Li W, Pang Y, Jin K, Wang Y, Wu Y, Luo J, Xu W, Zhang X, Xu R, Wang T, Jiao L. Membrane contact sites orchestrate cholesterol homeostasis that is central to vascular aging. WIREs Mech Dis 2023; 15:e1612. [PMID: 37156598 DOI: 10.1002/wsbm.1612] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2022] [Revised: 02/12/2023] [Accepted: 04/19/2023] [Indexed: 05/10/2023]
Abstract
Chronological age causes structural and functional vascular deterioration and is a well-established risk factor for the development of cardiovascular diseases, leading to more than 40% of all deaths in the elderly. The etiology of vascular aging is complex; a significant impact arises from impaired cholesterol homeostasis. Cholesterol level is balanced through synthesis, uptake, transport, and esterification, the processes executed by multiple organelles. Moreover, organelles responsible for cholesterol homeostasis are spatially and functionally coordinated instead of isolated by forming the membrane contact sites. Membrane contact, mediated by specific protein-protein interaction, pulls opposing organelles together and creates the hybrid place for cholesterol transfer and further signaling. The membrane contact-dependent cholesterol transfer, together with the vesicular transport, maintains cholesterol homeostasis and has intimate implications in a growing list of diseases, including vascular aging-related diseases. Here, we summarized the latest advances regarding cholesterol homeostasis by highlighting the membrane contact-based regulatory mechanism. We also describe the downstream signaling under cholesterol homeostasis perturbations, prominently in cholesterol-rich conditions, stimulating age-dependent organelle dysfunction and vascular aging. Finally, we discuss potential cholesterol-targeting strategies for therapists regarding vascular aging-related diseases. This article is categorized under: Cardiovascular Diseases > Molecular and Cellular Physiology.
Collapse
Affiliation(s)
- Wenjing Li
- Laboratory of Computational Biology and Machine Intelligence, National Laboratory of Pattern Recognition, Institute of Automation, Chinese Academy of Sciences, Beijing, China
- School of Artificial Intelligence, University of Chinese Academy of Sciences, Beijing, China
| | - Yiyun Pang
- Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Kehan Jin
- Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Yuru Wang
- Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Yujie Wu
- Laboratory of Computational Biology and Machine Intelligence, National Laboratory of Pattern Recognition, Institute of Automation, Chinese Academy of Sciences, Beijing, China
| | - Jichang Luo
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, Beijing, China
- China International Neuroscience Institute (China-INI), Beijing, China
| | - Wenlong Xu
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, Beijing, China
- China International Neuroscience Institute (China-INI), Beijing, China
| | - Xiao Zhang
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, Beijing, China
- China International Neuroscience Institute (China-INI), Beijing, China
| | - Ran Xu
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, Beijing, China
- China International Neuroscience Institute (China-INI), Beijing, China
| | - Tao Wang
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, Beijing, China
- China International Neuroscience Institute (China-INI), Beijing, China
| | - Liqun Jiao
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, Beijing, China
- China International Neuroscience Institute (China-INI), Beijing, China
- Department of Interventional Radiology, Xuanwu Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
31
|
Pérez-Luz S, Lalchandani J, Matamala N, Barrero MJ, Gil-Martín S, Saz SRD, Varona S, Monzón S, Cuesta I, Justo I, Marcacuzco A, Hierro L, Garfia C, Gomez-Mariano G, Janciauskiene S, Martínez-Delgado B. Quantitative Lipid Profiling Reveals Major Differences between Liver Organoids with Normal Pi*M and Deficient Pi*Z Variants of Alpha-1-antitrypsin. Int J Mol Sci 2023; 24:12472. [PMID: 37569847 PMCID: PMC10419530 DOI: 10.3390/ijms241512472] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Revised: 07/27/2023] [Accepted: 08/03/2023] [Indexed: 08/13/2023] Open
Abstract
Different mutations in the SERPINA1 gene result in alpha-1 antitrypsin (AAT) deficiency and in an increased risk for the development of liver diseases. More than 90% of severe deficiency patients are homozygous for Z (Glu342Lys) mutation. This mutation causes Z-AAT polymerization and intrahepatic accumulation which can result in hepatic alterations leading to steatosis, fibrosis, cirrhosis, and/or hepatocarcinoma. We aimed to investigate lipid status in hepatocytes carrying Z and normal M alleles of the SERPINA1 gene. Hepatic organoids were developed to investigate lipid alterations. Lipid accumulation in HepG2 cells overexpressing Z-AAT, as well as in patient-derived hepatic organoids from Pi*MZ and Pi*ZZ individuals, was evaluated by Oil-Red staining in comparison to HepG2 cells expressing M-AAT and liver organoids from Pi*MM controls. Furthermore, mass spectrometry-based lipidomics analysis and transcriptomic profiling were assessed in Pi*MZ and Pi*ZZ organoids. HepG2 cells expressing Z-AAT and liver organoids from Pi*MZ and Pi*ZZ patients showed intracellular accumulation of AAT and high numbers of lipid droplets. These latter paralleled with augmented intrahepatic lipids, and in particular altered proportion of triglycerides, cholesterol esters, and cardiolipins. According to transcriptomic analysis, Pi*ZZ organoids possess many alterations in genes and cellular processes of lipid metabolism with a specific impact on the endoplasmic reticulum, mitochondria, and peroxisome dysfunction. Our data reveal a relationship between intrahepatic accumulation of Z-AAT and alterations in lipid homeostasis, which implies that liver organoids provide an excellent model to study liver diseases related to the mutation of the SERPINA1 gene.
Collapse
Affiliation(s)
- Sara Pérez-Luz
- Molecular Genetics and Genetic Diagnostic Units, Institute of Rare Diseases Research (IIER), Spanish National Institute of Health Carlos III (ISCIII), 28220 Madrid, Spain; (S.P.-L.); (J.L.); (N.M.); (S.G.-M.); (S.R.-D.S.); (G.G.-M.)
| | - Jaanam Lalchandani
- Molecular Genetics and Genetic Diagnostic Units, Institute of Rare Diseases Research (IIER), Spanish National Institute of Health Carlos III (ISCIII), 28220 Madrid, Spain; (S.P.-L.); (J.L.); (N.M.); (S.G.-M.); (S.R.-D.S.); (G.G.-M.)
| | - Nerea Matamala
- Molecular Genetics and Genetic Diagnostic Units, Institute of Rare Diseases Research (IIER), Spanish National Institute of Health Carlos III (ISCIII), 28220 Madrid, Spain; (S.P.-L.); (J.L.); (N.M.); (S.G.-M.); (S.R.-D.S.); (G.G.-M.)
| | - Maria Jose Barrero
- Models and Mechanisms Unit, Institute of Rare Diseases Research (IIER), Spanish National Institute of Health Carlos III (ISCIII), 28220 Madrid, Spain;
| | - Sara Gil-Martín
- Molecular Genetics and Genetic Diagnostic Units, Institute of Rare Diseases Research (IIER), Spanish National Institute of Health Carlos III (ISCIII), 28220 Madrid, Spain; (S.P.-L.); (J.L.); (N.M.); (S.G.-M.); (S.R.-D.S.); (G.G.-M.)
- Centro de Investigación Biomédica en Red de Enfermedades Raras, CIBERER U758, 28029 Madrid, Spain
| | - Sheila Ramos-Del Saz
- Molecular Genetics and Genetic Diagnostic Units, Institute of Rare Diseases Research (IIER), Spanish National Institute of Health Carlos III (ISCIII), 28220 Madrid, Spain; (S.P.-L.); (J.L.); (N.M.); (S.G.-M.); (S.R.-D.S.); (G.G.-M.)
| | - Sarai Varona
- Bioinformatics Unit, Institute of Health Carlos III (ISCIII), 28220 Madrid, Spain; (S.V.); (S.M.); (I.C.)
| | - Sara Monzón
- Bioinformatics Unit, Institute of Health Carlos III (ISCIII), 28220 Madrid, Spain; (S.V.); (S.M.); (I.C.)
| | - Isabel Cuesta
- Bioinformatics Unit, Institute of Health Carlos III (ISCIII), 28220 Madrid, Spain; (S.V.); (S.M.); (I.C.)
| | - Iago Justo
- General and Digestive Surgery Department, Hospital 12 de Octubre, 28041 Madrid, Spain; (I.J.); (A.M.)
| | - Alberto Marcacuzco
- General and Digestive Surgery Department, Hospital 12 de Octubre, 28041 Madrid, Spain; (I.J.); (A.M.)
| | - Loreto Hierro
- Paediatric Hepatology Service, Research Institute of University Hospital La Paz, (IdiPAZ), 28046 Madrid, Spain;
| | - Cristina Garfia
- Digestive Department, Hospital 12 de Octubre, 28041 Madrid, Spain;
| | - Gema Gomez-Mariano
- Molecular Genetics and Genetic Diagnostic Units, Institute of Rare Diseases Research (IIER), Spanish National Institute of Health Carlos III (ISCIII), 28220 Madrid, Spain; (S.P.-L.); (J.L.); (N.M.); (S.G.-M.); (S.R.-D.S.); (G.G.-M.)
| | - Sabina Janciauskiene
- Department of Respiratory Medicine, Member of the German Center for Lung Research (DZL), Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), Hannover Medical School, 30625 Hannover, Germany;
| | - Beatriz Martínez-Delgado
- Molecular Genetics and Genetic Diagnostic Units, Institute of Rare Diseases Research (IIER), Spanish National Institute of Health Carlos III (ISCIII), 28220 Madrid, Spain; (S.P.-L.); (J.L.); (N.M.); (S.G.-M.); (S.R.-D.S.); (G.G.-M.)
- Centro de Investigación Biomédica en Red de Enfermedades Raras, CIBERER U758, 28029 Madrid, Spain
| |
Collapse
|
32
|
Perez-Luz S, Matamala N, Gomez-Mariano G, Janciauskiene S, Martínez-Delgado B. NAFLD and AATD Are Two Diseases with Unbalanced Lipid Metabolism: Similarities and Differences. Biomedicines 2023; 11:1961. [PMID: 37509601 PMCID: PMC10377048 DOI: 10.3390/biomedicines11071961] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Revised: 07/04/2023] [Accepted: 07/07/2023] [Indexed: 07/30/2023] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) is a type of steatosis commonly associated with obesity, dyslipidemia, hypertension, and diabetes. Other diseases such as inherited alpha-1 antitrypsin deficiency (AATD) have also been related to the development of liver steatosis. The primary reasons leading to hepatic lipid deposits can be genetic and epigenetic, and the outcomes range from benign steatosis to liver failure, as well as to extrahepatic diseases. Progressive hepatocellular damage and dysregulated systemic immune responses can affect extrahepatic organs, specifically the heart and lungs. In this review, we discuss the similarities and differences between the molecular pathways of NAFLD and AATD, and the putative value of hepatic organoids as novel models to investigate the physio pathological mechanisms of liver steatosis.
Collapse
Affiliation(s)
- Sara Perez-Luz
- Molecular Genetics Unit, Institute of Rare Diseases Research (IIER), Institute of Health Carlos III (ISCIII), 28220 Madrid, Spain
| | - Nerea Matamala
- Molecular Genetics Unit, Institute of Rare Diseases Research (IIER), Institute of Health Carlos III (ISCIII), 28220 Madrid, Spain
| | - Gema Gomez-Mariano
- Molecular Genetics Unit, Institute of Rare Diseases Research (IIER), Institute of Health Carlos III (ISCIII), 28220 Madrid, Spain
| | - Sabina Janciauskiene
- Department of Respiratory Medicine and Infectious Diseases, Biomedical Research in Endstage and Obstructive Lung Disease Hannover BREATH, Member of the German Center for Lung Research DZL, Hannover Medical School, 30625 Hannover, Germany
| | - Beatriz Martínez-Delgado
- Molecular Genetics Unit, Institute of Rare Diseases Research (IIER), Institute of Health Carlos III (ISCIII), 28220 Madrid, Spain
- CIBER de Enfermedades Raras, Instituto de Salud Carlos III, CIBERER U758, 28029 Madrid, Spain
| |
Collapse
|
33
|
Rivero-Pino F, Villanueva Á, Montserrat-de-la-Paz S, Sanchez-Fidalgo S, Millán-Linares MC. Evidence of Immunomodulatory Food-Protein Derived Peptides in Human Nutritional Interventions: Review on the Outcomes and Potential Limitations. Nutrients 2023; 15:2681. [PMID: 37375585 DOI: 10.3390/nu15122681] [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: 05/23/2023] [Revised: 06/01/2023] [Accepted: 06/07/2023] [Indexed: 06/29/2023] Open
Abstract
The immune system is somehow related to all the metabolic pathways, in a bidirectional way, and the nutritional interventions affecting these pathways might have a relevant impact on the inflammatory status of the individuals. Food-derived peptides have been demonstrated to exert several bioactivities by in vitro or animal studies. Their potential to be used as functional food is promising, considering the simplicity of their production and the high value of the products obtained. However, the number of human studies performed until now to demonstrate effects in vivo is still scarce. Several factors must be taken into consideration to carry out a high-quality human study to demonstrate immunomodulatory-promoting properties of a test item. This review aims to summarize the recent human studies published in which the purpose was to demonstrate bioactivity of protein hydrolysates, highlighting the main results and the limitations that can restrict the relevance of the studies. Results collected are promising, although in some studies, physiological changes could not be observed. When responses were observed, they sometimes did not refer to relevant parameters and the immunomodulatory properties could not be clearly established with the current evidence. Well-designed clinical trials are needed in order to evaluate the role of protein hydrolysates in immunonutrition.
Collapse
Affiliation(s)
- Fernando Rivero-Pino
- Department of Medical Biochemistry, Molecular Biology, and Immunology, School of Medicine, University of Seville, Av. Sanchez Pizjuan s/n, 41009 Seville, Spain
| | - Álvaro Villanueva
- Department of Food & Health, Instituto de la Grasa (IG-CSIC), Campus Universitario Pablo de Olavide, Ctra. Utrera Km. 1, 41013 Seville, Spain
| | - Sergio Montserrat-de-la-Paz
- Department of Medical Biochemistry, Molecular Biology, and Immunology, School of Medicine, University of Seville, Av. Sanchez Pizjuan s/n, 41009 Seville, Spain
| | - Susana Sanchez-Fidalgo
- Department of Preventive Medicine and Public Health, School of Medicine, University of Seville, Av. Sanchez Pizjuan s/n, 41009 Seville, Spain
| | - Maria C Millán-Linares
- Department of Medical Biochemistry, Molecular Biology, and Immunology, School of Medicine, University of Seville, Av. Sanchez Pizjuan s/n, 41009 Seville, Spain
| |
Collapse
|
34
|
Ouyang B, Deng L, Yang F, Shi H, Wang N, Tang W, Huang X, Zhou Y, Yu H, Wei Y, Dong J. Albumin-based formononetin nanomedicines for lung injury and fibrosis therapy via blocking macrophage pyroptosis. Mater Today Bio 2023; 20:100643. [PMID: 37214555 PMCID: PMC10193015 DOI: 10.1016/j.mtbio.2023.100643] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2023] [Revised: 03/23/2023] [Accepted: 04/23/2023] [Indexed: 05/24/2023] Open
Abstract
Pulmonary fibrosis that occurs following lung injury is a progressive and fatal disease since continual damage to lung tissue triggers the dysregulated inflammation response and accompanying abnormal healing process. Pyroptosis of alveolar macrophages has been found to play an essential role in the deterioration of lung injury and fibrosis. However, the lack of inhibitors against this inflammatory cell death in macrophages and the dense stroma pose major barriers to lung injury and fibrosis treatment. Herein, we developed an albumin-based nanoformulation to realize active delivery of formononetin (FMN) to improve the treatment of lung injury and fibrosis. The obtained nanoparticle, FMN@BSA NPs, could efficiently accumulate at the impaired lesion benefiting from the leaky vasculatures and the affinity between albumin and the overexpressed SPARC protein. Through blocking the NLRP3 inflammasome-involved pyroptosis process of macrophages, FMN@BSA NPs remarkably improved lung function and prolonged animal survival in the bleomycin (BLM)-induced lung injury and fibrosis model without noticeable side effects. Meanwhile, we proved FMN as a pyroptosis inhibitor and the corresponding lipid metabolism-related mechanisms through multi-omics analysis. This study first employed an albumin-based nanoparticle to deliver the pyroptosis inhibitor to the impaired lung tissue actively, providing a promising strategy for lung injury and fibrosis treatment.
Collapse
Affiliation(s)
- Boshu Ouyang
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai, 200040, PR China
- Institutes of Integrative Medicine, Fudan University, Shanghai, 200040, PR China
| | - Lingling Deng
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai, 200040, PR China
- Institutes of Integrative Medicine, Fudan University, Shanghai, 200040, PR China
| | - Fangyong Yang
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai, 200040, PR China
- Institutes of Integrative Medicine, Fudan University, Shanghai, 200040, PR China
| | - Hanlin Shi
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai, 200040, PR China
- Institutes of Integrative Medicine, Fudan University, Shanghai, 200040, PR China
| | - Na Wang
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai, 200040, PR China
- Institutes of Integrative Medicine, Fudan University, Shanghai, 200040, PR China
| | - Weifeng Tang
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai, 200040, PR China
- Institutes of Integrative Medicine, Fudan University, Shanghai, 200040, PR China
| | - Xi Huang
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai, 200040, PR China
- Institutes of Integrative Medicine, Fudan University, Shanghai, 200040, PR China
| | - Yaolong Zhou
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai, 200040, PR China
- Institutes of Integrative Medicine, Fudan University, Shanghai, 200040, PR China
| | - Hang Yu
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai, 200040, PR China
- Institutes of Integrative Medicine, Fudan University, Shanghai, 200040, PR China
| | - Ying Wei
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai, 200040, PR China
- Institutes of Integrative Medicine, Fudan University, Shanghai, 200040, PR China
| | - Jingcheng Dong
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai, 200040, PR China
- Institutes of Integrative Medicine, Fudan University, Shanghai, 200040, PR China
| |
Collapse
|
35
|
Iacobazzi D, Convertini P, Todisco S, Santarsiero A, Iacobazzi V, Infantino V. New Insights into NF-κB Signaling in Innate Immunity: Focus on Immunometabolic Crosstalks. BIOLOGY 2023; 12:776. [PMID: 37372061 DOI: 10.3390/biology12060776] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Revised: 05/23/2023] [Accepted: 05/24/2023] [Indexed: 06/29/2023]
Abstract
The nuclear factor kappa B (NF-κB) is a family of transcription factors that, beyond their numberless functions in various cell processes, play a pivotal role in regulating immune cell activation. Two main pathways-canonical and non-canonical-are responsible for NF-κB activation and heterodimer translocation into the nucleus. A complex crosstalk between NF-κB signaling and metabolism is emerging in innate immunity. Metabolic enzymes and metabolites regulate NF-κB activity in many cases through post-translational modifications such as acetylation and phosphorylation. On the other hand, NF-κB affects immunometabolic pathways, including the citrate pathway, thereby building an intricate network. In this review, the emerging findings about NF-κB function in innate immunity and the interplay between NF-κB and immunometabolism have been discussed. These outcomes allow for a deeper comprehension of the molecular mechanisms underlying NF-κB function in innate immune cells. Moreover, the new insights are important in order to perceive NF-κB signaling as a potential therapeutic target for inflammatory/immune chronic diseases.
Collapse
Affiliation(s)
- Dominga Iacobazzi
- Bristol Medical School, Translational Health Sciences, University of Bristol, Bristol BS2 8HW, UK
| | - Paolo Convertini
- Department of Science, University of Basilicata, Viale dell'Ateneo Lucano 10, 85100 Potenza, Italy
| | - Simona Todisco
- Department of Science, University of Basilicata, Viale dell'Ateneo Lucano 10, 85100 Potenza, Italy
| | - Anna Santarsiero
- Department of Science, University of Basilicata, Viale dell'Ateneo Lucano 10, 85100 Potenza, Italy
| | - Vito Iacobazzi
- Department of Biosciences, Biotechnologies and Biopharmaceutics, University of Bari, Via Orabona 4, 70125 Bari, Italy
| | - Vittoria Infantino
- Department of Science, University of Basilicata, Viale dell'Ateneo Lucano 10, 85100 Potenza, Italy
| |
Collapse
|
36
|
Favilli L, Griffith CM, Schymanski EL, Linster CL. High-throughput Saccharomyces cerevisiae cultivation method for credentialing-based untargeted metabolomics. Anal Bioanal Chem 2023:10.1007/s00216-023-04724-5. [PMID: 37212869 DOI: 10.1007/s00216-023-04724-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Revised: 04/24/2023] [Accepted: 04/28/2023] [Indexed: 05/23/2023]
Abstract
Identifying metabolites in model organisms is critical for many areas of biology, including unravelling disease aetiology or elucidating functions of putative enzymes. Even now, hundreds of predicted metabolic genes in Saccharomyces cerevisiae remain uncharacterized, indicating that our understanding of metabolism is far from complete even in well-characterized organisms. While untargeted high-resolution mass spectrometry (HRMS) enables the detection of thousands of features per analysis, many of these have a non-biological origin. Stable isotope labelling (SIL) approaches can serve as credentialing strategies to distinguish biologically relevant features from background signals, but implementing these experiments at large scale remains challenging. Here, we developed a SIL-based approach for high-throughput untargeted metabolomics in S. cerevisiae, including deep-48 well format-based cultivation and metabolite extraction, building on the peak annotation and verification engine (PAVE) tool. Aqueous and nonpolar extracts were analysed using HILIC and RP liquid chromatography, respectively, coupled to Orbitrap Q Exactive HF mass spectrometry. Of the approximately 37,000 total detected features, only 3-7% of the features were credentialed and used for data analysis with open-source software such as MS-DIAL, MetFrag, Shinyscreen, SIRIUS CSI:FingerID, and MetaboAnalyst, leading to the successful annotation of 198 metabolites using MS2 database matching. Comparable metabolic profiles were observed for wild-type and sdh1Δ yeast strains grown in deep-48 well plates versus the classical shake flask format, including the expected increase in intracellular succinate concentration in the sdh1Δ strain. The described approach enables high-throughput yeast cultivation and credentialing-based untargeted metabolomics, providing a means to efficiently perform molecular phenotypic screens and help complete metabolic networks.
Collapse
Affiliation(s)
- Lorenzo Favilli
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, Avenue du Swing 6, Belvaux, L-4367, Luxembourg.
| | - Corey M Griffith
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, Avenue du Swing 6, Belvaux, L-4367, Luxembourg
| | - Emma L Schymanski
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, Avenue du Swing 6, Belvaux, L-4367, Luxembourg
| | - Carole L Linster
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, Avenue du Swing 6, Belvaux, L-4367, Luxembourg
| |
Collapse
|
37
|
Yang W, Ma L, Xu S, Zheng P, Du J, Wu J, Yu J, Sun T. Gentiopicroside alleviated epileptogenesis in immature rats through inactivation of NLRP3 inflammasome by inhibiting P2X7R expression. Int J Dev Neurosci 2023; 83:53-66. [PMID: 36342791 DOI: 10.1002/jdn.10237] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2021] [Revised: 10/30/2022] [Accepted: 11/03/2022] [Indexed: 11/09/2022] Open
Abstract
OBJECTIVES This study aimed to elucidate the effects of Gentiopicroside (Gent) on epileptogenesis and underlying mechanisms. METHODS The status epilepticus (SE) model was established by intraperitoneal (i.p.) injection of lithium chloride (127 mg/kg) and pilocarpine (50 mg/kg) in immature rats. HAPI microglial cellular inflammation model was induced by lipopolysaccharide (LPS, 1 μg/ml) and adenosine triphosphate (ATP, 5 mM). The differential concentrations of Gent were used to pretreat animal (200, 400, and 800 mg/kg) and model cells (50, 100, and 200 μM). Epileptic discharges were assessed by electroencephalography (EEG) and Racine scale. Changes in spatial memory function were measured using the Morris water maze task test. Nissl and FJB staining were employed to assess the damage to hippocampus tissues. ELISA was used to detect the production of IL-1β, IL-18, and TNF-α. The expressions of P2X7R and NLRP3 were detected by q-PCR, immunofluorescence staining, and Western blot, and cell viability was determined by cell counting kit-8 (CCK-8). RESULTS Lithium chloride and pilocarpine (LICL-PILO) induced abnormal EEG activities, behavioral alterations, brain damage, and inflammatory responses in immature rats. However, Gent pretreatment significantly reduced the neuronal damage and spatial memory dysfunction induced by LICL-PILO. Additionally, Gent suppressed the production of inflammatory cytokines and inhibited the expression of P2X7R, NLRP3, ASC, and Caspase-1 in LPS/ATP-induced HAPI microglial cells. DISCUSSION Gent intervention could improve epileptogenesis in immature rats partially due to suppressing P2X7R and NLRP3 inflammasome.
Collapse
Affiliation(s)
- Weilong Yang
- School of Clinical Medicine, Ningxia Medical University, Yinchuan, China.,Department of Neurosurgery, The First Affiliated Hospital of Xinxiang Medical University, Wei Hui, China
| | - Lin Ma
- Department of Pharmacology, Ningxia Medical University, Yinchuan, China
| | - Siying Xu
- School of Clinical Medicine, Ningxia Medical University, Yinchuan, China
| | - Ping Zheng
- Department of Pharmacology, Ningxia Medical University, Yinchuan, China
| | - Juan Du
- Department of Pharmacology, Ningxia Medical University, Yinchuan, China
| | - Jing Wu
- Laboratory Animal Centre, Ningxia Medical University, Yinchuan, China
| | - Jianqiang Yu
- Department of Pharmacology, Ningxia Medical University, Yinchuan, China
| | - Tao Sun
- Ningxia Key Laboratory of Craniocerebral Diseases of Ningxia Hui Autonomous Region, Ningxia Medical University, Yinchuan, China
| |
Collapse
|
38
|
Olona A, Leishman S, Anand PK. The NLRP3 inflammasome: regulation by metabolic signals. Trends Immunol 2022; 43:978-989. [PMID: 36371361 DOI: 10.1016/j.it.2022.10.003] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Revised: 10/05/2022] [Accepted: 10/08/2022] [Indexed: 11/10/2022]
Abstract
Macrophages undergo profound metabolic reprogramming upon sensing infectious and sterile stimuli. This metabolic shift supports and regulates essential innate immune functions, including activation of the NLRP3 inflammasome. Within distinct metabolic networks, key enzymes play pivotal roles to control flux restraining detrimental inflammasome signaling. However, depending on the metabolic cues, specific enzymes and metabolites result in inflammasome activation outcomes which contrast other metabolic steps in the pathway. We posit that understanding which metabolic steps commit to discrete inflammasome fates will broaden our understanding of metabolic checkpoints to maintain homeostasis and offer better therapeutic options in human disease.
Collapse
Affiliation(s)
- Antoni Olona
- Department of Infectious Disease, Imperial College London, London, W12 0NN, UK; Program in Cardiovascular and Metabolic Disorders, and Centre for Computational Biology, Duke-NUS Medical School, Singapore
| | - Stuart Leishman
- Department of Infectious Disease, Imperial College London, London, W12 0NN, UK
| | - Paras K Anand
- Department of Infectious Disease, Imperial College London, London, W12 0NN, UK.
| |
Collapse
|
39
|
Zhang J, Kong X, Sun W, Wang L, Shen T, Chen M, Chen X. The RNA-binding protein RBM24 regulates lipid metabolism and SLC7A11 mRNA stability to modulate ferroptosis and inflammatory response. Front Cell Dev Biol 2022; 10:1008576. [PMID: 36478739 PMCID: PMC9720322 DOI: 10.3389/fcell.2022.1008576] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Accepted: 11/04/2022] [Indexed: 11/22/2022] Open
Abstract
Lipids play a critical role in many cellular processes by serving as structural components of cell membranes or functioning as energy fuel and signaling molecules. The RNA-binding proteins RBM24 and RBM38 share an identical RNA-binding domain and thereby, regulate a group of same targets, such as p21. However, it is not certain whether RBM24 and RBM38 participates in lipid homeostasis. Here, lipidomic analysis showed that a deficiency in RBM24 or RBM38 leads to altered lipid metabolism, with more profound alteration by loss of RBM24 in MCF7 cells. We also showed that mice deficient in RBM24 were prone to chronic inflammation and liver steatosis, but not spontaneous tumors. These data let us speculate whether RBM24 regulates ferroptosis, a programmed cell death that links inflammation and liver steatosis via lipid peroxidation. Indeed, we found that over-expression of RBM24 protected, whereas knockout of RBM24 sensitized, cells to Erastin-induced ferroptosis by modulating the mRNA stability of SLC7A11, a ferroptosis inhibitor. Moreover, we showed that knockdown of SLC7A11 reversed the effect of RBM24 on ferroptosis. Together, our study revealed that RBM24 regulates lipid metabolism and SLC7A11 mRNA stability to modulate ferroptosis and inflammatory response.
Collapse
Affiliation(s)
- Jin Zhang
- Comparative Oncology Laboratory, Schools of Veterinary Medicine and Medicine, UC, Davis, CA, United States,*Correspondence: Jin Zhang, ; Xinbin Chen,
| | - Xiangmudong Kong
- Comparative Oncology Laboratory, Schools of Veterinary Medicine and Medicine, UC, Davis, CA, United States
| | | | - Leyi Wang
- Comparative Oncology Laboratory, Schools of Veterinary Medicine and Medicine, UC, Davis, CA, United States
| | - Tong Shen
- West Coast Metabolomics Center, UC, Davis, CA, United States
| | - Mingyi Chen
- Department of Pathology, Southwestern Medical Center, University of Texas, Austin, TX, United States
| | - Xinbin Chen
- Comparative Oncology Laboratory, Schools of Veterinary Medicine and Medicine, UC, Davis, CA, United States,*Correspondence: Jin Zhang, ; Xinbin Chen,
| |
Collapse
|
40
|
The Hypolipidemic Effect of Hawthorn Leaf Flavonoids through Modulating Lipid Metabolism and Gut Microbiota in Hyperlipidemic Rats. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2022; 2022:3033311. [PMID: 36425260 PMCID: PMC9681556 DOI: 10.1155/2022/3033311] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Revised: 10/28/2022] [Accepted: 10/29/2022] [Indexed: 11/16/2022]
Abstract
Objective. The purpose of this study was to explore the potential mechanisms of the lipid-regulating effects and the effect on modulating the gut microbiota of hawthorn leaf flavonoids (HLF) in the high-fat diet-induced hyperlipidemic rats. Methods. The hypolipidemic effect of HLF was investigated in the high-fat diet-induced hyperlipidemic rats. The action targets of HLF in the treatment of hyperlipidemia were predicted by network pharmacology and KEGG enrichment bubble diagram, which were verified by the test of western blotting. Meanwhile, we used 16S rRNA sequencing to evaluate the effects of HLF on the microbes. Results. The results of animal experiments showed that HLF could reduce the body weight and regulate the levels of serum lipid in high-fat diet (HFD) rats. Meanwhile, for the related targets of cholesterol metabolism, HLF could significantly upregulate the expression of LDLR, NR1H3, and ABCG5/ABCG8; reduce the expression of PCSK9; and increase the level of CYP7A1 in the intestinal tissue, whereas cholesterol biosynthetic protein expressions including HMGCR and SCAP were lowered by HLF. In addition, HLF increased the activities of plasma SOD, CAT, and GSH-Px and decreased the levels of Casp 1, NLRP3, IL-1β, IL-18, and TNF-α, improving the degree of hepatocyte steatosis and inflammatory infiltration of rats. Notably, HLF significantly regulated the relative abundance of major bacteria such as g_Lactobacillus, g_Anaerostipes, g_[Eubacterium]_hallii_group, g_Fusicatenibacter, g_Akkermansia, and g_Collinsella. Synchronously, we found that HLF could regulate the disorder of plasma HEPC and TFR levels caused by HFD. Conclusion. This study demonstrates that HLF can regulate metabolic hyperlipidemia syndromes and modulate the relative abundance of major bacteria, which illustrated that it might be associated with the modulation of gut microbiota composition and metabolites.
Collapse
|
41
|
Verbeke R, Hogan MJ, Loré K, Pardi N. Innate immune mechanisms of mRNA vaccines. Immunity 2022; 55:1993-2005. [PMID: 36351374 PMCID: PMC9641982 DOI: 10.1016/j.immuni.2022.10.014] [Citation(s) in RCA: 172] [Impact Index Per Article: 57.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Revised: 09/08/2022] [Accepted: 10/13/2022] [Indexed: 11/09/2022]
Abstract
The lipid nanoparticle (LNP)-encapsulated, nucleoside-modified mRNA platform has been used to generate safe and effective vaccines in record time against COVID-19. Here, we review the current understanding of the manner whereby mRNA vaccines induce innate immune activation and how this contributes to protective immunity. We discuss innate immune sensing of mRNA vaccines at the cellular and intracellular levels and consider the contribution of both the mRNA and the LNP components to their immunogenicity. A key message that is emerging from recent observations is that the LNP carrier acts as a powerful adjuvant for this novel vaccine platform. In this context, we highlight important gaps in understanding and discuss how new insight into the mechanisms underlying the effectiveness of mRNA-LNP vaccines may enable tailoring mRNA and carrier molecules to develop vaccines with greater effectiveness and milder adverse events in the future.
Collapse
Affiliation(s)
- Rein Verbeke
- Ghent Research Group on Nanomedicines, Faculty of Pharmacy, Ghent University, Ottergemsesteenweg 460, 9000 Ghent, Belgium; Department of Biochemistry and Molecular Biology, University of British Columbia, BC V6T 1Z4, Vancouver, Canada.
| | - Michael J Hogan
- Department of Pathology and Laboratory Medicine, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Karin Loré
- Division of Immunology and Allergy, Department of Medicine Solna, Karolinska Institutet, Karolinska University Hospital, 171 64 Solna, Sweden; Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Norbert Pardi
- Department of Microbiology, University of Pennsylvania, Philadelphia, PA 19104, USA.
| |
Collapse
|
42
|
Fang J, Zhang Y, Chen D, Zheng Y, Jiang J. Exosomes and Exosomal Cargos: A Promising World for Ventricular Remodeling Following Myocardial Infarction. Int J Nanomedicine 2022; 17:4699-4719. [PMID: 36217495 PMCID: PMC9547598 DOI: 10.2147/ijn.s377479] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2022] [Accepted: 09/21/2022] [Indexed: 11/23/2022] Open
Abstract
Exosomes are a pluripotent group of extracellular nanovesicles secreted by all cells that mediate intercellular communications. The effective information within exosomes is primarily reflected in exosomal cargos, including proteins, lipids, DNAs, and non-coding RNAs (ncRNAs), the most intensively studied molecules. Cardiac resident cells (cardiomyocytes, fibroblasts, and endothelial cells) and foreign cells (infiltrated immune cells, cardiac progenitor cells, cardiosphere-derived cells, and mesenchymal stem cells) are involved in the progress of ventricular remodeling (VR) following myocardial infarction (MI) via transferring exosomes into target cells. Here, we summarize the pathological mechanisms of VR following MI, including cardiac myocyte hypertrophy, cardiac fibrosis, inflammation, pyroptosis, apoptosis, autophagy, angiogenesis, and metabolic disorders, and the roles of exosomal cargos in these processes, with a focus on proteins and ncRNAs. Continued research in this field reveals a novel diagnostic and therapeutic strategy for VR.
Collapse
Affiliation(s)
- Jiacheng Fang
- Department of Cardiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310009, People’s Republic of China
| | - Yuxuan Zhang
- Department of Cardiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310009, People’s Republic of China
| | - Delong Chen
- Department of Cardiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310009, People’s Republic of China
| | - Yiyue Zheng
- Department of Cardiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310009, People’s Republic of China
| | - Jun Jiang
- Department of Cardiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310009, People’s Republic of China,Correspondence: Jun Jiang, Department of Cardiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, No. 88 Jiefang Road, Hangzhou, Zhejiang, 310009, People’s Republic of China, Tel/Fax +86 135 8870 6891, Email
| |
Collapse
|
43
|
Capece D, Verzella D, Flati I, Arboretto P, Cornice J, Franzoso G. NF-κB: blending metabolism, immunity, and inflammation. Trends Immunol 2022; 43:757-775. [PMID: 35965153 DOI: 10.1016/j.it.2022.07.004] [Citation(s) in RCA: 236] [Impact Index Per Article: 78.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Revised: 07/11/2022] [Accepted: 07/12/2022] [Indexed: 02/06/2023]
Abstract
The procurement and management of nutrients and ability to fight infections are fundamental requirements for survival. These defense responses are bioenergetically costly, requiring the immune system to balance protection against pathogens with the need to maintain metabolic homeostasis. NF-κB transcription factors are central regulators of immunity and inflammation. Over the last two decades, these factors have emerged as a pivotal node coordinating the immune and metabolic systems in physiology and the etiopathogenesis of major threats to human health, including cancer, autoimmunity, chronic inflammation, and others. In this review, we discuss recent advances in understanding how NF-κB-dependent metabolic programs control inflammation, metabolism, and immunity and how improved knowledge of them may lead to better diagnostics and therapeutics for widespread human diseases.
Collapse
Affiliation(s)
- Daria Capece
- Department of Biotechnological and Applied Clinical Sciences (DISCAB), University of L'Aquila, 67100 L'Aquila, Italy; Department of Immunology and Inflammation, Imperial College London, London W12 0NN, UK.
| | - Daniela Verzella
- Department of Biotechnological and Applied Clinical Sciences (DISCAB), University of L'Aquila, 67100 L'Aquila, Italy; Department of Immunology and Inflammation, Imperial College London, London W12 0NN, UK
| | - Irene Flati
- Department of Biotechnological and Applied Clinical Sciences (DISCAB), University of L'Aquila, 67100 L'Aquila, Italy
| | - Paola Arboretto
- Department of Immunology and Inflammation, Imperial College London, London W12 0NN, UK
| | - Jessica Cornice
- Department of Immunology and Inflammation, Imperial College London, London W12 0NN, UK
| | - Guido Franzoso
- Department of Immunology and Inflammation, Imperial College London, London W12 0NN, UK.
| |
Collapse
|
44
|
Liu T, Lu X, Gao W, Zhai Y, Li H, Li S, Yang L, Ma F, Zhan Y, Mao H. Cardioprotection effect of Yiqi-Huoxue-Jiangzhuo formula in a chronic kidney disease mouse model associated with gut microbiota modulation and NLRP3 inflammasome inhibition. Biomed Pharmacother 2022; 152:113159. [PMID: 35661533 DOI: 10.1016/j.biopha.2022.113159] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Revised: 05/15/2022] [Accepted: 05/16/2022] [Indexed: 11/29/2022] Open
Abstract
BACKGROUND The pathogenesis and treatment of cardiovascular disease mediated by chronic kidney disease (CKD) are key research questions. Specifically, the mechanisms underlying the cardiorenal protective effect of Yiqi-Huoxue-Jiangzhuo formula (YHJF), a traditional Chinese herbal medicine, have not yet been clarified. METHODS A classical CKD mouse model was constructed by 5/6 nephrectomy (Nx) to study the effects of YHJF intervention on 5/6 Nx mice cardiorenal function, gut microbial composition, gut-derived metabolites, and NLRP3 inflammasome pathways. RESULTS YHJF improved cardiac dysfunction and reversed left ventricular hypertrophy, myocardial hypertrophy, and interstitial fibrosis in 5/6 Nx mice. In addition, YHJF inhibited activation of the NLRP3 inflammasome and downregulated the expression of TNF-α and IL-1β both in the heart and serum; reconstitution of the intestinal flora imbalance was also found in 5/6 Nx mice treated with YHJF. Spearman's correlation and redundancy analyses showed that changes in the intestinal flora of 5/6 Nx mice were related to clinical phenotype and serum inflammatory levels. CONCLUSIONS Treatment with YHJF effectively protected the heart function of 5/6 Nx mice; this effect was attributed to inhibition of NLRP3 inflammasome activation and regulation of intestinal microbial composition and derived metabolites. YHJF has potential for improving intestinal flora imbalance and gut-derived toxin accumulation in patients with CKD, thereby preventing cardiovascular complications.
Collapse
Affiliation(s)
- Tongtong Liu
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China
| | - Xiaoguang Lu
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China
| | - Wenya Gao
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Yuanyuan Zhai
- China-Japan Friendship Hospital, Institute of Medical Science, Beijing 100029, China
| | - Han Li
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China
| | - Shangheng Li
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China
| | - Liping Yang
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China
| | - Fang Ma
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China
| | - Yongli Zhan
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China.
| | - Huimin Mao
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China.
| |
Collapse
|
45
|
Findeisen HM, Voges VC, Braun LC, Sonnenberg J, Schwarz D, Körner H, Reinecke H, Sohrabi Y. LXRα Regulates oxLDL-Induced Trained Immunity in Macrophages. Int J Mol Sci 2022; 23:ijms23116166. [PMID: 35682840 PMCID: PMC9181299 DOI: 10.3390/ijms23116166] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Revised: 05/26/2022] [Accepted: 05/26/2022] [Indexed: 12/19/2022] Open
Abstract
Reprogramming of metabolic pathways in monocytes and macrophages can induce a proatherosclerotic inflammatory memory called trained innate immunity. Here, we have analyzed the role of the Liver X receptor (LXR), a crucial regulator of metabolism and inflammation, in oxidized low-density lipoprotein (oxLDL)-induced trained innate immunity. Human monocytes were incubated with LXR agonists, antagonists, and oxLDL for 24 h. After five days of resting time, cells were restimulated with the TLR-2 agonist Pam3cys. OxLDL priming induced the expression of LXRα but not LXRβ. Pharmacologic LXR activation was enhanced, while LXR inhibition prevented the oxLDL-induced inflammatory response. Furthermore, LXR inhibition blocked the metabolic changes necessary for epigenetic reprogramming associated with trained immunity. In fact, enrichment of activating histone marks at the IL-6 and TNFα promotor was reduced following LXR inhibition. Based on the differential expression of the LXR isoforms, we inhibited LXRα and LXRβ genes using siRNA in THP1 cells. As expected, siRNA-mediated knock-down of LXRα blocked the oxLDL-induced inflammatory response, while knock-down of LXRβ had no effect. We demonstrate a specific and novel role of the LXRα isoform in the regulation of oxLDL-induced trained immunity. Our data reveal important aspects of LXR signaling in innate immunity with relevance to atherosclerosis formation.
Collapse
|
46
|
Challagundla N, Saha B, Agrawal-Rajput R. Insights into inflammasome regulation: cellular, molecular, and pathogenic control of inflammasome activation. Immunol Res 2022; 70:578-606. [PMID: 35610534 DOI: 10.1007/s12026-022-09286-9] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Accepted: 05/04/2022] [Indexed: 02/07/2023]
Abstract
Maintenance of immune homeostasis is an intricate process wherein inflammasomes play a pivotal role by contributing to innate and adaptive immune responses. Inflammasomes are ensembles of adaptor proteins that can trigger a signal following innate sensing of pathogens or non-pathogens eventuating in the inductions of IL-1β and IL-18. These inflammatory cytokines substantially influence the antigen-presenting cell's costimulatory functions and T helper cell differentiation, contributing to adaptive immunity. As acute and chronic disease conditions may accompany parallel tissue damage, we analyze the critical role of extracellular factors such as cytokines, amyloids, cholesterol crystals, etc., intracellular metabolites, and signaling molecules regulating inflammasome activation/inhibition. We develop an operative framework for inflammasome function and regulation by host cell factors and pathogens. While inflammasomes influence the innate and adaptive immune components' interplay modulating the anti-pathogen adaptive immune response, pathogens may target inflammasome inhibition as a survival strategy. As trapped between health and diseases, inflammasomes serve as promising therapeutic targets and their modus operandi serves as a scientific rationale for devising better therapeutic strategies.
Collapse
Affiliation(s)
- Naveen Challagundla
- Immunology lab, Indian Institute of Advanced Research, Gandhinagar, Gujarat, 382007, India
| | - Bhaskar Saha
- National Centre for Cell Science, Lab-5, Ganeshkhind, Pune, Maharashtra, 411007, India
| | - Reena Agrawal-Rajput
- Immunology lab, Indian Institute of Advanced Research, Gandhinagar, Gujarat, 382007, India.
| |
Collapse
|
47
|
Disulfiram inhibits oxidative stress and NLRP3 inflammasome activation to prevent LPS-induced cardiac injury. Int Immunopharmacol 2022; 105:108545. [DOI: 10.1016/j.intimp.2022.108545] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2021] [Revised: 01/10/2022] [Accepted: 01/12/2022] [Indexed: 12/18/2022]
|
48
|
Phytotherapeutic Approaches to the Prevention of Age-Related Changes and the Extension of Active Longevity. Molecules 2022; 27:molecules27072276. [PMID: 35408672 PMCID: PMC9000830 DOI: 10.3390/molecules27072276] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Revised: 03/29/2022] [Accepted: 03/30/2022] [Indexed: 02/01/2023] Open
Abstract
Maintaining quality of life with an increase in life expectancy is considered one of the global problems of our time. This review explores the possibility of using natural plant compounds with antioxidant, anti-inflammatory, anti-glycation, and anti-neurodegenerative properties to slow down the onset of age-related changes. Age-related changes such as a decrease in mental abilities, the development of inflammatory processes, and increased risk of developing type 2 diabetes have a significant impact on maintaining quality of life. Herbal preparations can play an essential role in preventing and treating neurodegenerative diseases that accompany age-related changes, including Alzheimer’s and Parkinson’s diseases. Medicinal plants have known sedative, muscle relaxant, neuroprotective, nootropic, and antiparkinsonian properties. The secondary metabolites, mainly polyphenolic compounds, are valuable substances for the development of new anti-inflammatory and hypoglycemic agents. Understanding how mixtures of plants and their biologically active substances work together to achieve a specific biological effect can help develop targeted drugs to prevent diseases associated with aging and age-related changes. Understanding the mechanisms of the biological activity of plant complexes and mixtures determines the prospects for using metabolomic and biochemical methods to prolong active longevity.
Collapse
|
49
|
Li H, Guo Z, Chen J, Du Z, Lu H, Wang Z, Xi J, Bai Y. Computational research of Belnacasan and new Caspase-1 inhibitor on cerebral ischemia reperfusion injury. Aging (Albany NY) 2022; 14:1848-1864. [PMID: 35193116 PMCID: PMC8908936 DOI: 10.18632/aging.203907] [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: 11/10/2021] [Accepted: 01/25/2022] [Indexed: 11/25/2022]
Abstract
Cerebral ischemia-reperfusion injury is one of the most severe diseases in terms of mortality and disability, which seriously threatens human life and health. In clinical treatment, drug thrombolysis or mechanical interventional thrombolysis are used to quickly restore the blood supply of ischemic brain tissue. But with the rapid recovery of blood flow, complex pathophysiological processes such as oxidative stress and inflammation will further aggravate brain tissue damage, namely cerebral ischemia-reperfusion injury, for which there is no effective treatment. Recent studies have shown that the medical community has paid the role of inflammation and pyroptosis in cerebral ischemia-reperfusion injury more and more attention. And Caspase-1 was found to play a vital role in regulating inflammation pathways and pyroptosis in many inflammation-associated diseases, especially in cerebral ischemia-reperfusion injury. Not only that, Caspase-1 inhibitors have been shown to reduce the damage of cerebral ischemia-reperfusion injury by inhibiting inflammation and pyroptosis. And the Caspase-1 inhibitor, Belnacasan, has been proved to modify the active site of Caspase-1 and lead to the blocking of Caspase-1, thus correlating with tissue protection of inflammatory diseases in animal models. Therefore, it’s essential to screen and design potential Caspase-1 inhibitors to reduce cerebral ischemia-reperfusion injury and protect brain function by reducing inflammation and pyroptosis, which provides a new idea for clinical treatment of the cerebral ischemia-reperfusion injury. This study applied a group of computer-aided technology, such as Discovery Studio 4.5, Schrodinger, and PyMol, to screen and assess potential Caspase-1 inhibitors. Moreover, the ADME (absorption, distribution, metabolism, excretion) and TOPKAT (Toxicity Prediction by Computer Assisted Technology) molecules of Discovery Studio 4.5 were conducted to evaluate molecules' pharmacological and toxicological features. Then, precise molecular docking was applied to assess the binding mechanism and affinity between Caspase-1 and selected compounds. Besides, molecular dynamics simulations were performed to determine the stability of ligand-receptor complexes in the natural environment. In summary, this study lists promising drug candidates and their pharmacological properties, promoting the development of Caspase-1 inhibitors and deepening the understanding of the interaction between inhibitors and Caspase-1.
Collapse
Affiliation(s)
- Hui Li
- Department of Neurology, The First Hospital of Jilin University, Changchun, China
| | - Zhen Guo
- Clinical College, Jilin University, Changchun, China
| | - Jun Chen
- Clinical College, Jilin University, Changchun, China
| | - Zhishan Du
- Clinical College, Jilin University, Changchun, China
| | - Han Lu
- Clinical College, Jilin University, Changchun, China
| | - Zhenhua Wang
- Clinical College, Jilin University, Changchun, China
| | - Jianxin Xi
- Clinical College, Jilin University, Changchun, China
| | - Yang Bai
- Department of Neurosurgery, The First Hospital of Jilin University, Changchun, China
| |
Collapse
|
50
|
Krämer J, Kang R, Grimm LM, De Cola L, Picchetti P, Biedermann F. Molecular Probes, Chemosensors, and Nanosensors for Optical Detection of Biorelevant Molecules and Ions in Aqueous Media and Biofluids. Chem Rev 2022; 122:3459-3636. [PMID: 34995461 PMCID: PMC8832467 DOI: 10.1021/acs.chemrev.1c00746] [Citation(s) in RCA: 162] [Impact Index Per Article: 54.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Indexed: 02/08/2023]
Abstract
Synthetic molecular probes, chemosensors, and nanosensors used in combination with innovative assay protocols hold great potential for the development of robust, low-cost, and fast-responding sensors that are applicable in biofluids (urine, blood, and saliva). Particularly, the development of sensors for metabolites, neurotransmitters, drugs, and inorganic ions is highly desirable due to a lack of suitable biosensors. In addition, the monitoring and analysis of metabolic and signaling networks in cells and organisms by optical probes and chemosensors is becoming increasingly important in molecular biology and medicine. Thus, new perspectives for personalized diagnostics, theranostics, and biochemical/medical research will be unlocked when standing limitations of artificial binders and receptors are overcome. In this review, we survey synthetic sensing systems that have promising (future) application potential for the detection of small molecules, cations, and anions in aqueous media and biofluids. Special attention was given to sensing systems that provide a readily measurable optical signal through dynamic covalent chemistry, supramolecular host-guest interactions, or nanoparticles featuring plasmonic effects. This review shall also enable the reader to evaluate the current performance of molecular probes, chemosensors, and nanosensors in terms of sensitivity and selectivity with respect to practical requirement, and thereby inspiring new ideas for the development of further advanced systems.
Collapse
Affiliation(s)
- Joana Krämer
- Institute
of Nanotechnology, Karlsruhe Institute of
Technology (KIT), Hermann-von-Helmholtz Platz 1, 76344 Eggenstein-Leopoldshafen, Germany
| | - Rui Kang
- Institute
of Nanotechnology, Karlsruhe Institute of
Technology (KIT), Hermann-von-Helmholtz Platz 1, 76344 Eggenstein-Leopoldshafen, Germany
| | - Laura M. Grimm
- Institute
of Nanotechnology, Karlsruhe Institute of
Technology (KIT), Hermann-von-Helmholtz Platz 1, 76344 Eggenstein-Leopoldshafen, Germany
| | - Luisa De Cola
- Institute
of Nanotechnology, Karlsruhe Institute of
Technology (KIT), Hermann-von-Helmholtz Platz 1, 76344 Eggenstein-Leopoldshafen, Germany
- Dipartimento
DISFARM, University of Milano, via Camillo Golgi 19, 20133 Milano, Italy
- Department
of Molecular Biochemistry and Pharmacology, Instituto di Ricerche Farmacologiche Mario Negri, IRCCS, 20156 Milano, Italy
| | - Pierre Picchetti
- Institute
of Nanotechnology, Karlsruhe Institute of
Technology (KIT), Hermann-von-Helmholtz Platz 1, 76344 Eggenstein-Leopoldshafen, Germany
| | - Frank Biedermann
- Institute
of Nanotechnology, Karlsruhe Institute of
Technology (KIT), Hermann-von-Helmholtz Platz 1, 76344 Eggenstein-Leopoldshafen, Germany
| |
Collapse
|