101
|
Personalized Nutrition in the Management of Female Infertility: New Insights on Chronic Low-Grade Inflammation. Nutrients 2022; 14:nu14091918. [PMID: 35565885 PMCID: PMC9105997 DOI: 10.3390/nu14091918] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Revised: 04/19/2022] [Accepted: 04/29/2022] [Indexed: 02/01/2023] Open
Abstract
Increasing evidence on the significance of nutrition in reproduction is emerging from both animal and human studies, suggesting a mutual association between nutrition and female fertility. Different “fertile” dietary patterns have been studied; however, in humans, conflicting results or weak correlations are often reported, probably because of the individual variations in genome, proteome, metabolome, and microbiome and the extent of exposure to different environmental conditions. In this scenario, “precision nutrition”, namely personalized dietary patterns based on deep phenotyping and on metabolomics, microbiome, and nutrigenetics of each case, might be more efficient for infertile patients than applying a generic nutritional approach. In this review, we report on new insights into the nutritional management of infertile patients, discussing the main nutrigenetic, nutrigenomic, and microbiomic aspects that should be investigated to achieve effective personalized nutritional interventions. Specifically, we will focus on the management of low-grade chronic inflammation, which is associated with several infertility-related diseases.
Collapse
|
102
|
Huang M, Xiao M, Dong J, Huang Y, Sun H, Wang D. Synergistic anti-inflammatory effects of graphene oxide quantum dots and trans-10-hydroxy-2-decenoic acid on LPS-stimulated RAW 264.7 macrophage cells. BIOMATERIALS ADVANCES 2022; 136:212774. [PMID: 35929313 DOI: 10.1016/j.bioadv.2022.212774] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/28/2021] [Revised: 03/14/2022] [Accepted: 03/20/2022] [Indexed: 06/15/2023]
Abstract
Graphene oxide quantum dots (GOQDs) have attracted substantial attention in numerous fields due to their unique physicochemical properties. However, their nanotoxicity and potential for use in biomedicine still require further study. In this work, the effects of GOQD and trans- 10-hydroxy-2-decenoic acid (10-HDA) cotreatment on the immune function of macrophages (RAW264.7 cells) were investigated. In particular, LC/MS-based metabolomics was performed to evaluate the effects of GOQDs on the metabolism of LPS-stimulated macrophages. Herein, we fabricated GOQDs with good dispersibility and a uniform size distribution of approximately 7 nm using a polyimide-pyrolyzed carbon film as the working electrode, a high-voltage graphite electrode as the cathode, and H2O2 as the oxidant. The GOQDs entered the macrophages and emitted green fluorescence under UV irradiation. Cotreatment with GOQDs and 10-HDA induced RAW 264.7 cell proliferation. GOQDs promoted the anti-inflammatory effect of 10-HDA on LPS-stimulated RAW264.7 cells and attenuated the secretion of TNF-α, IL-6, and IL-1β. The metabolites in RAW264.7 cells treated with GOQDs were significantly different from those in RAW264.7 cells treated with LPS. The enrichment analysis showed that treatment with GOQDs interfered with amino acid metabolism, and lipid metabolism. Our results demonstrate the role of GOQDs in macrophages and provide a basis for their further application in biomedical fields.
Collapse
Affiliation(s)
- Minjie Huang
- Institute of Animal Husbandry and Veterinary Science, Zhejiang Academy of Agricultural Sciences, 145 Shiqiao Road, Hangzhou 310021, China
| | - Minhui Xiao
- Institute of Animal Husbandry and Veterinary Science, Zhejiang Academy of Agricultural Sciences, 145 Shiqiao Road, Hangzhou 310021, China
| | - Jie Dong
- Institute of Animal Husbandry and Veterinary Science, Zhejiang Academy of Agricultural Sciences, 145 Shiqiao Road, Hangzhou 310021, China
| | - Yee Huang
- Institute of Animal Husbandry and Veterinary Science, Zhejiang Academy of Agricultural Sciences, 145 Shiqiao Road, Hangzhou 310021, China
| | - Haiyan Sun
- Hangzhou Gaoxi Technol Co Ltd, 6 Naxian Street, Hangzhou 310013, Zhejiang, China.
| | - Deqian Wang
- Institute of Animal Husbandry and Veterinary Science, Zhejiang Academy of Agricultural Sciences, 145 Shiqiao Road, Hangzhou 310021, China.
| |
Collapse
|
103
|
Scott NA, Lawson MAE, Hodgetts RJ, Le Gall G, Hall LJ, Mann ER. Macrophage metabolism in the intestine is compartment specific and regulated by the microbiota. Immunology 2022; 166:138-152. [PMID: 35199335 DOI: 10.1111/imm.13461] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2021] [Revised: 01/31/2022] [Accepted: 02/17/2022] [Indexed: 11/28/2022] Open
Abstract
Intestinal macrophages play a vital role in the maintenance of gut homeostasis through signals derived from the microbiota. We previously demonstrated that microbial-derived metabolites can shape the metabolic functions of macrophages. Here, we show that antibiotic-induced disruption of the intestinal microbiota dramatically alters both the local metabolite environment and the metabolic functions of macrophages in the colon. Broad-spectrum antibiotic administration in mice increased the expression of the large neutral amino acid transporter LAT1 and accordingly, amino acid uptake. Subsequently, antibiotic administration enhanced the metabolic functions of colonic macrophages, increasing phosphorylation of components of mammalian/mechanistic target of rapamycin signalling pathways, with increased expression of genes involved in glycolysis and oxidative phosphorylation (OXPHOS), increased mitochondrial function, increased rate of extracellular acidification (ECAR; measure of glycolysis) and increased rate of oxygen consumption (OCR; measure of OXPHOS). Small bowel macrophages were less metabolically active than their colonic counterparts, with macrophage metabolism in the small intestine being independent of the microbiota. Finally, we reveal tissue-resident Tim4+ CD4+ macrophages exhibit enhanced fatty acid uptake alongside reduced fatty acid synthesis compared to recruited macrophages. Thus, the microbiota shapes gut macrophage metabolism in a compartment-specific manner, with important implications for monocyte recruitment and macrophage differentiation.
Collapse
Affiliation(s)
- Nicholas A Scott
- Division of Infection, Immunity and Respiratory Medicine, Faculty of Biology, Medicine and Health, School of Biological Sciences, University of Manchester, Manchester Academic Health Science Centre, Lydia Becker Institute of Immunology and Inflammation, Manchester, UK
| | - Melissa A E Lawson
- Division of Infection, Immunity and Respiratory Medicine, Faculty of Biology, Medicine and Health, School of Biological Sciences, University of Manchester, Manchester Academic Health Science Centre, Lydia Becker Institute of Immunology and Inflammation, Manchester, UK.,Gut Microbes & Health, Quadram Institute Bioscience, Norwich Research Park, Norwich, UK
| | - Ryan James Hodgetts
- Division of Infection, Immunity and Respiratory Medicine, Faculty of Biology, Medicine and Health, School of Biological Sciences, University of Manchester, Manchester Academic Health Science Centre, Lydia Becker Institute of Immunology and Inflammation, Manchester, UK.,Department of Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - Gwénaëlle Le Gall
- Norwich Medical School, University of East Anglia, Norwich Research Park, Norwich, UK
| | - Lindsay J Hall
- Gut Microbes & Health, Quadram Institute Bioscience, Norwich Research Park, Norwich, UK.,ZIEL - Institute for Food & Health, Technical University of Munich, Freising, Germany
| | - Elizabeth R Mann
- Division of Infection, Immunity and Respiratory Medicine, Faculty of Biology, Medicine and Health, School of Biological Sciences, University of Manchester, Manchester Academic Health Science Centre, Lydia Becker Institute of Immunology and Inflammation, Manchester, UK
| |
Collapse
|
104
|
Entezari M, Sadrkhanloo M, Rashidi M, Asnaf SE, Taheriazam A, Hashemi M, Ashrafizadeh M, Zarrabi A, Rabiee N, Hushmandi K, Mirzaei S, Sethi G. Non-coding RNAs and macrophage interaction in tumor progression. Crit Rev Oncol Hematol 2022; 173:103680. [PMID: 35405273 DOI: 10.1016/j.critrevonc.2022.103680] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Revised: 03/25/2022] [Accepted: 04/06/2022] [Indexed: 12/12/2022] Open
Abstract
The macrophages are abundantly found in TME and their M2 polarization is in favor of tumor malignancy. On the other hand, non-coding RNAs (ncRNAs) can modulate macrophage polarization in TME to affect cancer progression. The miRNAs can dually induce/suppress M2 polarization of macrophages and by affecting various molecular pathways, they modulate tumor progression and therapy response. The lncRNAs can affect miRNAs via sponging and other molecular pathways to modulate macrophage polarization. A few experiments have also examined role of circRNAs in targeting signaling networks and affecting macrophages. The therapeutic targeting of these ncRNAs can mediate TME remodeling and affect macrophage polarization. Furthermore, exosomal ncRNAs derived from tumor cells or macrophages can modulate polarization and TME remodeling. Suppressing biogenesis and secretion of exosomes can inhibit ncRNA-mediated M2 polarization of macrophages and prevent tumor progression. The ncRNAs, especially exosomal ncRNAs can be considered as non-invasive biomarkers for tumor diagnosis.
Collapse
Affiliation(s)
- Maliheh Entezari
- Department of Genetics, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran; Farhikhtegan Medical Convergence sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | | | - Mohsen Rashidi
- Department Pharmacology, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran; The Health of Plant and Livestock Products Research Center, Mazandaran University of Medical Sciences, Sari, Iran
| | - Sholeh Etehad Asnaf
- Department of Cell and Molecular Biology, Faculty of Biological Sciences, North Tehran Branch, Islamic Azad University, Tehran, Iran
| | - Afshin Taheriazam
- Farhikhtegan Medical Convergence sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran; Department of Orthopedics, Faculty of medicine, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Mehrdad Hashemi
- Department of Genetics, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran; Farhikhtegan Medical Convergence sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Milad Ashrafizadeh
- Faculty of Engineering and Natural Sciences, Sabanci University, Orta Mahalle, Üniversite Caddesi No. 27, Orhanlı, Tuzla, Istanbul, Turkey
| | - Ali Zarrabi
- Department of Biomedical Engineering, Faculty of Engineering and Natural Sciences, Istinye University, Istanbul 34396, Turkey
| | - Navid Rabiee
- School of Engineering, Macquarie University, Sydney, New South Wales 2109, Australia
| | - Kiavash Hushmandi
- Department of Food Hygiene and Quality Control, Division of Epidemiology & Zoonoses, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran.
| | - Sepideh Mirzaei
- Department of Biology, Faculty of Science, Islamic Azad University, Science and Research Branch, Tehran, Iran.
| | - Gautam Sethi
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore; Cancer Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore.
| |
Collapse
|
105
|
Chen H, Zhang Y, Li X, Zhang W, He H, Du B, Li T, Tang H, Liu Y, Li L, Shi M. Transcriptome Changes and Potential Immunotoxicity Analysis in RAW264.7 Macrophages Caused by Bisphenol F. Front Pharmacol 2022; 13:846562. [PMID: 35387338 PMCID: PMC8978606 DOI: 10.3389/fphar.2022.846562] [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: 12/31/2021] [Accepted: 02/15/2022] [Indexed: 11/17/2022] Open
Abstract
As a viable substitute for bisphenol A (BPA), BPF has been widely used in the plastic industry and daily consumer goods, resulting in its detection in humans at a comparable concentration. Evidence reveals that BPF and BPA may have similar toxic effects due to their similar structures. However, there is less information about BPF and its latent implications on the immune system, which is associated with many disorders. In this study, the in vitro toxicity of BPF on RAW264.7 macrophages was explored. The cells were treated with different concentrations of BPF (5, 10, 20, 50, 100, and 200 μM), the cell viability and apoptosis were detected, the gene expression profile was analyzed by whole-transcriptome sequencing, and the mRNA levels were detected by qRT-PCR. The results showed a high concentration of BPF could significantly reduce the survival rate of RAW264.7 macrophages. Although the medium concentration (20–50 μM) of BPF seemed to have no impact on the cell activity of macrophages, it caused the occurrence of apoptosis. The results of differential transcription showed that compared with the control group, 121 genes were upregulated and 82 genes were downregulated in the BPF group. The significantly changed gene functions were mainly concentrated in cell cycle, phagosome, lysosome, and antigen processing and presentation. These findings provide valuable information for correctly understanding the immunotoxicity risk of BPF and may help to improve the hazard identification of bisphenol compounds.
Collapse
Affiliation(s)
- Huiling Chen
- Dongguan Key Laboratory of Environmental Medicine, School of Public Health, Guangdong Medical University, Dongguan, China
| | - Yanchao Zhang
- Dongguan Key Laboratory of Environmental Medicine, School of Public Health, Guangdong Medical University, Dongguan, China
| | - Xing Li
- Dongguan Key Laboratory of Environmental Medicine, School of Public Health, Guangdong Medical University, Dongguan, China
| | - Wei Zhang
- Dongguan Key Laboratory of Environmental Medicine, School of Public Health, Guangdong Medical University, Dongguan, China
| | - Haoqi He
- Dongguan Key Laboratory of Environmental Medicine, School of Public Health, Guangdong Medical University, Dongguan, China
| | - Bohai Du
- Dongguan Key Laboratory of Environmental Medicine, School of Public Health, Guangdong Medical University, Dongguan, China
| | - Tianlan Li
- Dongguan Key Laboratory of Environmental Medicine, School of Public Health, Guangdong Medical University, Dongguan, China
| | - Huanwen Tang
- Dongguan Key Laboratory of Environmental Medicine, School of Public Health, Guangdong Medical University, Dongguan, China
| | - Yungang Liu
- Department of Toxicology, School of Public Health, Southern Medical University, Guangzhou, China
| | - Li Li
- Dongguan Key Laboratory of Environmental Medicine, School of Public Health, Guangdong Medical University, Dongguan, China
| | - Ming Shi
- Dongguan Key Laboratory of Environmental Medicine, School of Public Health, Guangdong Medical University, Dongguan, China.,Dongguan Liaobu Hospital, Dongguan, China
| |
Collapse
|
106
|
Impact of Lipid Metabolism on Antitumor Immune Response. Cancers (Basel) 2022; 14:cancers14071850. [PMID: 35406621 PMCID: PMC8997602 DOI: 10.3390/cancers14071850] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Revised: 04/01/2022] [Accepted: 04/05/2022] [Indexed: 02/04/2023] Open
Abstract
Simple Summary One of the causes of failure of anticancer therapies is the reprogramming of lipid metabolism. Cells of innate and adaptive immunity present in the tumor microenvironment can be affected by this metabolic switch and thus present changes in their anti- or protumor phenotype. In this review, modifications induced by lipid metabolism will be described for innate immune cells, such as macrophages, dendritic cells and MDSCs, and also for adaptive immune cells, such as CD4+ and CD8+ T cells and B cells. Finally, antitumor therapeutic strategies targeting lipid metabolism will be presented. Abstract Over the past decade, metabolic reprogramming has been defined as a hallmark of cancer. More recently, a large number of studies have demonstrated that metabolic reprogramming can modulate the differentiation and functions of immune cells, and thus modify the antitumor response. Increasing evidence suggests that modified energy metabolism could be responsible for the failure of antitumor immunity. Indeed, tumor-infiltrating immune cells play a key role in cancer, and metabolic switching in these cells has been shown to help determine their phenotype: tumor suppressive or immune suppressive. Recent studies in the field of immunometabolism focus on metabolic reprogramming in the tumor microenvironment (TME) by targeting innate and adaptive immune cells and their associated anti- or protumor phenotypes. In this review, we discuss the lipid metabolism of immune cells in the TME as well as the effects of lipids; finally, we expose the link between therapies and lipid metabolism.
Collapse
|
107
|
Mogilenko DA, Danko K, Larionova EE, Shavva VS, Kudriavtsev IV, Nekrasova EV, Burnusuz AV, Gorbunov NP, Trofimov AV, Zhakhov AV, Ivanov IA, Orlov SV. Differentiation of human macrophages with anaphylatoxin C3a impairs alternative M2 polarization and decreases lipopolysaccharide‐induced cytokine secretion. Immunol Cell Biol 2022; 100:186-204. [DOI: 10.1111/imcb.12534] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2021] [Revised: 01/06/2022] [Accepted: 02/07/2022] [Indexed: 11/28/2022]
Affiliation(s)
- Denis A Mogilenko
- Department of Biochemistry Institute of Experimental Medicine St. Petersburg Russia
- Department of Embryology St. Petersburg State University St. Petersburg Russia
| | - Katerina Danko
- Department of Biochemistry Institute of Experimental Medicine St. Petersburg Russia
- Department of Cytology and Histology St. Petersburg State University St. Petersburg Russia
| | | | - Vladimir S Shavva
- Department of Biochemistry Institute of Experimental Medicine St. Petersburg Russia
| | - Igor V Kudriavtsev
- Department of Cytology and Histology St. Petersburg State University St. Petersburg Russia
- Department of Immunology Institute of Experimental Medicine St. Petersburg Russia
| | | | - Alexandra V Burnusuz
- Department of Biochemistry Institute of Experimental Medicine St. Petersburg Russia
- Department of Cytology and Histology St. Petersburg State University St. Petersburg Russia
- Department of Immunology Institute of Experimental Medicine St. Petersburg Russia
| | - Nikolay P Gorbunov
- The Research Institute of Highly Pure Biopreparations St. Petersburg Russia
| | | | | | | | - Sergey V Orlov
- Department of Biochemistry Institute of Experimental Medicine St. Petersburg Russia
- Department of Embryology St. Petersburg State University St. Petersburg Russia
| |
Collapse
|
108
|
Dai M, Yang X, Yu Y, Pan W. Helminth and Host Crosstalk: New Insight Into Treatment of Obesity and Its Associated Metabolic Syndromes. Front Immunol 2022; 13:827486. [PMID: 35281054 PMCID: PMC8913526 DOI: 10.3389/fimmu.2022.827486] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Accepted: 02/02/2022] [Indexed: 12/16/2022] Open
Abstract
Obesity and its associated Metabolic Syndromes (Mets) represent a global epidemic health problem. Metabolic inflammation, lipid accumulation and insulin resistance contribute to the progression of these diseases, thereby becoming targets for drug development. Epidemiological data have showed that the rate of helminth infection negatively correlates with the incidence of obesity and Mets. Correspondingly, numerous animal experiments and a few of clinic trials in human demonstrate that helminth infection or its derived molecules can mitigate obesity and Mets via induction of macrophage M2 polarization, inhibition of adipogenesis, promotion of fat browning, and improvement of glucose tolerance, insulin resistance and metabolic inflammation. Interestingly, sporadic studies also uncover that several helminth infections can reshape gut microbiota of hosts, which is intimately implicated in the pathogenesis of obesity and Mets. Overall, these findings indicate that the crosstalk between helminth and hosts may be a novel direction for obesity and Mets therapy. The present article reviews the molecular mechanism of how helminth masters immunity and metabolism in obesity.
Collapse
Affiliation(s)
- Mengyu Dai
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogen Biology and Immunology, Xuzhou Medical University, Xuzhou, China
- The Second Clinical Medicine, Xuzhou Medical University, Xuzhou, China
- National Demonstration Center for Experimental Basic Medical Science Education (Xuzhou Medical University), Xuzhou, China
| | - Xiaoying Yang
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogen Biology and Immunology, Xuzhou Medical University, Xuzhou, China
| | - Yinghua Yu
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogen Biology and Immunology, Xuzhou Medical University, Xuzhou, China
- *Correspondence: Wei Pan, ; Yinghua Yu,
| | - Wei Pan
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogen Biology and Immunology, Xuzhou Medical University, Xuzhou, China
- *Correspondence: Wei Pan, ; Yinghua Yu,
| |
Collapse
|
109
|
Bin YL, Hu HS, Tian F, Wen ZH, Yang MF, Wu BH, Wang LS, Yao J, Li DF. Metabolic Reprogramming in Gastric Cancer: Trojan Horse Effect. Front Oncol 2022; 11:745209. [PMID: 35096565 PMCID: PMC8790521 DOI: 10.3389/fonc.2021.745209] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Accepted: 11/12/2021] [Indexed: 12/24/2022] Open
Abstract
Worldwide, gastric cancer (GC) represents the fifth most common cancer for incidence and the third leading cause of death in developed countries. Despite the development of combination chemotherapies, the survival rates of GC patients remain unsatisfactory. The reprogramming of energy metabolism is a hallmark of cancer, especially increased dependence on aerobic glycolysis. In the present review, we summarized current evidence on how metabolic reprogramming in GC targets the tumor microenvironment, modulates metabolic networks and overcomes drug resistance. Preclinical and clinical studies on the combination of metabolic reprogramming targeted agents and conventional chemotherapeutics or molecularly targeted treatments [including vascular endothelial growth factor receptor (VEGFR) and HER2] and the value of biomarkers are examined. This deeper understanding of the molecular mechanisms underlying successful pharmacological combinations is crucial in finding the best-personalized treatment regimens for cancer patients.
Collapse
Affiliation(s)
- Yu-Ling Bin
- Department of Rheumatology and Immunology, ZhuZhou Central Hospital, Zhuzhou, China
| | - Hong-Sai Hu
- Department of Gastroenterology, ZhuZhou Central Hospital, Zhuzhou, China
| | - Feng Tian
- Department of Rheumatology and Immunology, ZhuZhou Central Hospital, Zhuzhou, China
| | - Zhen-Hua Wen
- Department of Rheumatology and Immunology, ZhuZhou Central Hospital, Zhuzhou, China
| | - Mei-Feng Yang
- Department of Hematology, Yantian District People's Hospital, Shenzhen, China
| | - Ben-Hua Wu
- Department of Gastroenterology, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University, The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, China
| | - Li-Sheng Wang
- Department of Gastroenterology, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University, The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, China
| | - Jun Yao
- Department of Gastroenterology, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University, The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, China
| | - De-Feng Li
- Department of Gastroenterology, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University, The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, China
| |
Collapse
|
110
|
Human placental mesenchymal stromal cell therapy restores the cytokine efflux and insulin signaling in the skeletal muscle of obesity-induced type 2 diabetes rat model. Hum Cell 2022; 35:557-571. [PMID: 35091972 DOI: 10.1007/s13577-021-00664-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Accepted: 12/14/2021] [Indexed: 11/04/2022]
Abstract
Obesity poses a significant risk factor for the onset of metabolic syndrome with allied complications, wherein mesenchymal stem cell therapy is seen as a promising treatment for obesity-induced metabolic syndrome. In the present study, we aim to explore the beneficial effects of the human placental mesenchymal stromal cells (P-MSCs) on obesity-associated insulin resistance (IR) including inflammation. To understand this, we have analyzed the peripheral blood glucose, serum insulin levels by ELISA, and the glucose uptake capacity of skeletal muscle by a 2-NBDG assay using flow cytometry in WNIN/GR-Ob rats treated with and without P-MSCs. Also, we have studied insulin signaling and cytokine profile in the skeletal muscle by western blotting, dot blotting, and Multiplex-ELISA techniques. The skeletal muscle of WNIN/GR-Ob rats demonstrates dysregulation of cytokines, altered glucose uptake vis-a-vis insulin signaling. However, P-MSCs' treatment was effective in WNIN/GR-Ob rats as compared to its control, to restore HOMA-IR, re-establishes dysregulated cytokines and PI3K-Akt pathway in addition to enhanced Glut4 expression and glucose uptake studied in skeletal muscle. Overall, our data advocate the beneficial effects of P-MSCs to ameliorate inflammatory milieu, improve insulin sensitivity, and normalize glucose homeostasis underlining the Ob-T2D conditions, and we attribute for immunomodulatory, paracrine, autocrine, and multipotent functions of P-MSCs.
Collapse
|
111
|
Zhang B, Xue Y, Zhao J, Jiang H, Zhu J, Yin H, Qiu Y, Hu A, Xu L, Song Y, Wang X. Shionone Attenuates Sepsis-Induced Acute Kidney Injury by Regulating Macrophage Polarization via the ECM1/STAT5 Pathway. Front Med (Lausanne) 2022; 8:796743. [PMID: 35141243 PMCID: PMC8818860 DOI: 10.3389/fmed.2021.796743] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2021] [Accepted: 12/28/2021] [Indexed: 12/16/2022] Open
Abstract
Backgrounds To date, there are no specific drugs approved for the treatment of sepsis associated acute kidney injury (AKI). Shionone is a natural component with anti-inflammatory activity. In this study, we sought to determine the functional role of Shionone in sepsis-induced AKI. Methods Animal models of AKI were constructed by cecum ligation and puncture (CLP) surgery. C57BL/6 mice were randomly assigned to the Sham, CLP, 10 mg/kg DXM, 50 mg/kg Shionone and 100 mg/kg Shionone groups. RAW264.7 treated with lipopolysaccharides (LPS) was used as an in vitro sepsis model and cells were divided into control, LPS, 1 μg/mL Shionone and 2 μg/mL Shionone groups. The pathological status was assessed by Hematoxylin-Eosin (HE) staining assay, protein expressions were detected by immunofluorescence staining and Western blot, macrophage typing was detected by flow, and the levels of pro-inflammatory factors (IL-6, IL-12, IL-1β, TNF-α) and anti-inflammatory factors (IL-10 and TGF-β) were measured using the corresponding kits. Results ECM1 is highly expressed in tissue-infiltrating macrophages under inflammatory conditions. It has been observed that Shionone inhibits the expression of ECM1 and attenuates sepsis-induced injury in kidney and inflammatory factor levels in serum. In addition, Shionone may reduce inflammatory factor levels through the promotion of M2 macrophages by GM-CSF/STAT5/Arg1 pathway to alleviate sepsis induced inflammation in vitro. Conclusion These findings demonstrate that Shionone can alleviate sepsis-induced AKI by promoting M2 macrophage polarization through regulating the ECM1/STAT5 pathway.
Collapse
Affiliation(s)
- Biao Zhang
- Department of Critical Care Medicine, Suzhou Hospital of Integrated Traditional Chinese and Western Medicine, Suzhou, China
| | - Yi Xue
- Department of Nephrology, Suzhou Hospital of Integrated Traditional Chinese and Western Medicine, Suzhou, China
| | - Jin Zhao
- Department of Critical Care Medicine, Suzhou Hospital of Integrated Traditional Chinese and Western Medicine, Suzhou, China
| | - Huojun Jiang
- Department of Critical Care Medicine, Suzhou Hospital of Integrated Traditional Chinese and Western Medicine, Suzhou, China
| | - Jiaoli Zhu
- Department of Nephrology, Suzhou Hospital of Integrated Traditional Chinese and Western Medicine, Suzhou, China
| | - Hao Yin
- Li Shicai School Inheritance Studio, Suzhou Hospital of Integrated Traditional and Western Medicine, Suzhou, China
| | - Yizhen Qiu
- Department of Critical Care Medicine, Suzhou Hospital of Integrated Traditional Chinese and Western Medicine, Suzhou, China
| | - Aihao Hu
- Department of Critical Care Medicine, Suzhou Hospital of Integrated Traditional Chinese and Western Medicine, Suzhou, China
| | - Lingqi Xu
- Department of Critical Care Medicine, Suzhou Hospital of Integrated Traditional Chinese and Western Medicine, Suzhou, China
| | - Yi Song
- Department of Critical Care Medicine, Suzhou Hospital of Integrated Traditional Chinese and Western Medicine, Suzhou, China
- Yi Song
| | - Xin Wang
- Li Shicai School Inheritance Studio, Suzhou Hospital of Integrated Traditional and Western Medicine, Suzhou, China
- *Correspondence: Xin Wang
| |
Collapse
|
112
|
Patsalos A, Halasz L, Medina-Serpas MA, Berger WK, Daniel B, Tzerpos P, Kiss M, Nagy G, Fischer C, Simandi Z, Varga T, Nagy L. A growth factor-expressing macrophage subpopulation orchestrates regenerative inflammation via GDF-15. J Exp Med 2022; 219:e20210420. [PMID: 34846534 PMCID: PMC8635277 DOI: 10.1084/jem.20210420] [Citation(s) in RCA: 47] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Revised: 08/03/2021] [Accepted: 11/01/2021] [Indexed: 12/13/2022] Open
Abstract
Muscle regeneration is the result of the concerted action of multiple cell types driven by the temporarily controlled phenotype switches of infiltrating monocyte-derived macrophages. Pro-inflammatory macrophages transition into a phenotype that drives tissue repair through the production of effectors such as growth factors. This orchestrated sequence of regenerative inflammatory events, which we termed regeneration-promoting program (RPP), is essential for proper repair. However, it is not well understood how specialized repair-macrophage identity develops in the RPP at the transcriptional level and how induced macrophage-derived factors coordinate tissue repair. Gene expression kinetics-based clustering of blood circulating Ly6Chigh, infiltrating inflammatory Ly6Chigh, and reparative Ly6Clow macrophages, isolated from injured muscle, identified the TGF-β superfamily member, GDF-15, as a component of the RPP. Myeloid GDF-15 is required for proper muscle regeneration following acute sterile injury, as revealed by gain- and loss-of-function studies. Mechanistically, GDF-15 acts both on proliferating myoblasts and on muscle-infiltrating myeloid cells. Epigenomic analyses of upstream regulators of Gdf15 expression identified that it is under the control of nuclear receptors RXR/PPARγ. Finally, immune single-cell RNA-seq profiling revealed that Gdf15 is coexpressed with other known muscle regeneration-associated growth factors, and their expression is limited to a unique subpopulation of repair-type macrophages (growth factor-expressing macrophages [GFEMs]).
Collapse
Affiliation(s)
- Andreas Patsalos
- Departments of Medicine and Biological Chemistry, Johns Hopkins University School of Medicine, Institute for Fundamental Biomedical Research, Johns Hopkins All Children’s Hospital, St. Petersburg, FL
| | - Laszlo Halasz
- Departments of Medicine and Biological Chemistry, Johns Hopkins University School of Medicine, Institute for Fundamental Biomedical Research, Johns Hopkins All Children’s Hospital, St. Petersburg, FL
| | - Miguel A. Medina-Serpas
- Departments of Medicine and Biological Chemistry, Johns Hopkins University School of Medicine, Institute for Fundamental Biomedical Research, Johns Hopkins All Children’s Hospital, St. Petersburg, FL
| | - Wilhelm K. Berger
- Departments of Medicine and Biological Chemistry, Johns Hopkins University School of Medicine, Institute for Fundamental Biomedical Research, Johns Hopkins All Children’s Hospital, St. Petersburg, FL
| | - Bence Daniel
- Departments of Medicine and Biological Chemistry, Johns Hopkins University School of Medicine, Institute for Fundamental Biomedical Research, Johns Hopkins All Children’s Hospital, St. Petersburg, FL
| | - Petros Tzerpos
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Máté Kiss
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Gergely Nagy
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | | | - Zoltan Simandi
- Sanford Burnham Prebys Medical Discovery Institute, Orlando, FL
| | - Tamas Varga
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Laszlo Nagy
- Departments of Medicine and Biological Chemistry, Johns Hopkins University School of Medicine, Institute for Fundamental Biomedical Research, Johns Hopkins All Children’s Hospital, St. Petersburg, FL
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| |
Collapse
|
113
|
Hu H, Mei J, Lin M, Wu X, Lin H, Chen G. The causal relationship between obesity and skin and soft tissue infections: A two-sample Mendelian randomization study. Front Endocrinol (Lausanne) 2022; 13:996863. [PMID: 36568121 PMCID: PMC9768473 DOI: 10.3389/fendo.2022.996863] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/18/2022] [Accepted: 11/22/2022] [Indexed: 12/12/2022] Open
Abstract
OBJECTIVE Many observational studies have shown that obesity strongly affects skin and soft tissue infections (SSTIs). However, whether a causal genetic relationship exists between obesity and SSTIs is unclear. METHODS A two-sample Mendelian randomization (MR) study was used to explore whether obesity is causally associated with SSTIs using a publicly released genome-wide association study (GWAS). An inverse-variance weighted (IVW) analysis was used as the primary analysis, and the results are reported as the odds ratios (ORs). Heterogeneity was tested using Cochran's Q test and the I2 statistic, and horizontal pleiotropy was tested using the MR-Egger intercept and MR pleiotropy residual sum and outlier (MR-PRESSO). RESULTS The results of the MR analysis showed a positive effect of BMI on SSTIs (OR 1.544, 95% CI 1.399-1.704, P= 5.86 × 10-18). After adjusting for the effect of type 2 diabetes (T2D) and peripheral vascular disease (PVD), the positive effect still existed. Then, we further assessed the effect of BMI on different types of SSTIs. The results showed that BMI caused an increased risk of impetigo, cutaneous abscess, furuncle and carbuncle, cellulitis, pilonidal cyst, and other local infections of skin and subcutaneous tissues, except for acute lymphadenitis. However, the associations disappeared after adjusting for the effect of T2D and PVD, and the associations between BMI and impetigo or cellulitis disappeared. Finally, we assessed the effects of several obesity-related characteristics on SSTIs. Waist circumference, hip circumference, body fat percentage, and whole-body fat mass, excluding waist-to-hip ratio, had a causal effect on an increased risk of SSTIs. However, the associations disappeared after adjusting for the effect of BMI. CONCLUSION This study found that obesity had a positive causal effect on SSTIs. Reasonable weight control is a possible way to reduce the occurrence of SSTIs, especially in patients undergoing surgery.
Collapse
Affiliation(s)
- Hongxin Hu
- Department of Orthopedic Surgery, Affiliated Hospital of Putian University, Putian, China
| | - Jian Mei
- Department of Orthopaedic Surgery, Experimental Orthopaedics, Centre for Medical Biotechnology (ZMB/Biopark 1), University of Regensburg, Regensburg, Germany
| | - Mei Lin
- Department of Surgery, Affiliated Hospital of Putian University, Putian, China
| | - Xianwei Wu
- Department of Orthopedic Surgery, Affiliated Hospital of Putian University, Putian, China
| | - Haibin Lin
- Department of Orthopedic Surgery, Affiliated Hospital of Putian University, Putian, China
| | - Guoli Chen
- Department of Orthopedic Surgery, Affiliated Hospital of Putian University, Putian, China
- *Correspondence: Guoli Chen,
| |
Collapse
|
114
|
Rathod S, Aggarwal V, Upadhyay A, Choudhari R. Timing of the Major Metabolic Switches in Immune Cell Activation and Differentiation During Cancer Development. IMMUNO-ONCOLOGY CROSSTALK AND METABOLISM 2022:187-218. [DOI: 10.1007/978-981-16-6226-3_7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
115
|
Alves RW, da Silva EM, Doretto-Silva L, Andrade-Oliveira V. Metabolic Pathways in Immune Cells Commitment and Fate. ESSENTIAL ASPECTS OF IMMUNOMETABOLISM IN HEALTH AND DISEASE 2022:53-82. [DOI: 10.1007/978-3-030-86684-6_4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
116
|
Chen Y, Li Y. Metabolic reprogramming and immunity in cancer. CANCER IMMUNOLOGY AND IMMUNOTHERAPY 2022:137-196. [DOI: 10.1016/b978-0-12-823397-9.00006-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
117
|
Dong H, Feng Y, Yang Y, Hu Y, Jia Y, Yang S, Zhao N, Zhao R. A Novel Function of Mitochondrial Phosphoenolpyruvate Carboxykinase as a Regulator of Inflammatory Response in Kupffer Cells. Front Cell Dev Biol 2022; 9:726931. [PMID: 34970539 PMCID: PMC8712867 DOI: 10.3389/fcell.2021.726931] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2021] [Accepted: 11/30/2021] [Indexed: 01/22/2023] Open
Abstract
Background: There has been a recent appreciation that some metabolic enzymes can profoundly influence the nature of the immune response produced in macrophages. However, the role of mitochondrial phosphoenolpyruvate carboxykinase (PCK2) in immune response remains unknown. This study aims to investigate the role of PCK2 in lipopolysaccharides (LPS)-induced activation in Kupffer cells. Methods: Inflammatory cytokines were determined by real-time quantitative reverse transcription-polymerase chain action (qRT-PCR) and flow cytometric analysis using a cytometric bead array. Western blotting and immunofluorescence staining were used to determine PCK2 expression and subcellular distribution under confocal laser microscopy. qRT-PCR, flow cytometry, and high-performance liquid chromatography (HPLC) were used to determine mitochondrial function. Pharmacological inhibition, knockdown, and overexpression of PCK2 were used to confirm its function. Co-immunoprecipitation (Co-IP) was performed to determine MAPK/NF-κB phosphorylation. Results: Inflammatory response was significantly increased in LPS-treated mice and Kupffer cells. During the inflammatory process, the protein level of PCK2 was significantly upregulated in Kupffer cells. Interestingly, the localization of PCK2 was mainly in cytosol rather than mitochondria after LPS stimulation. Gain-of-function and loss-of-function analyses found that PCK2 overexpression significantly upregulated the levels of inflammation markers, whereas PCK2 knockdown or inhibition significantly mitigated LPS-induced inflammatory response in Kupffer cells. Furthermore, PCK2 promoted protein phosphorylation of NF-κB and AKT/MAPK, the major signaling pathways, controlling inflammatory cascade activation. Conclusion: We identified a novel function of PCK2 in mediating LPS-induced inflammation and provided mechanistic insights into the regulation of inflammatory response in Kupffer cells. Therefore, PCK2 may serve as a novel therapeutic target for the regulation of Kupffer cells-mediated inflammatory responses.
Collapse
Affiliation(s)
- Haibo Dong
- MOE Joint International Research Laboratory of Animal Health and Food Safety, Nanjing Agricultural University, Nanjing, China.,Key Laboratory of Animal Physiology and Biochemistry, Nanjing Agricultural University, Nanjing, China
| | - Yue Feng
- MOE Joint International Research Laboratory of Animal Health and Food Safety, Nanjing Agricultural University, Nanjing, China.,Key Laboratory of Animal Physiology and Biochemistry, Nanjing Agricultural University, Nanjing, China
| | - Yang Yang
- MOE Joint International Research Laboratory of Animal Health and Food Safety, Nanjing Agricultural University, Nanjing, China.,Key Laboratory of Animal Physiology and Biochemistry, Nanjing Agricultural University, Nanjing, China
| | - Yun Hu
- MOE Joint International Research Laboratory of Animal Health and Food Safety, Nanjing Agricultural University, Nanjing, China.,Key Laboratory of Animal Physiology and Biochemistry, Nanjing Agricultural University, Nanjing, China
| | - Yimin Jia
- MOE Joint International Research Laboratory of Animal Health and Food Safety, Nanjing Agricultural University, Nanjing, China.,Key Laboratory of Animal Physiology and Biochemistry, Nanjing Agricultural University, Nanjing, China
| | - Shu Yang
- MOE Joint International Research Laboratory of Animal Health and Food Safety, Nanjing Agricultural University, Nanjing, China.,Key Laboratory of Animal Physiology and Biochemistry, Nanjing Agricultural University, Nanjing, China
| | - Nannan Zhao
- MOE Joint International Research Laboratory of Animal Health and Food Safety, Nanjing Agricultural University, Nanjing, China.,Key Laboratory of Animal Physiology and Biochemistry, Nanjing Agricultural University, Nanjing, China
| | - Ruqian Zhao
- MOE Joint International Research Laboratory of Animal Health and Food Safety, Nanjing Agricultural University, Nanjing, China.,Key Laboratory of Animal Physiology and Biochemistry, Nanjing Agricultural University, Nanjing, China
| |
Collapse
|
118
|
Xu L, Liu W, Bai F, Xu Y, Liang X, Ma C, Gao L. Hepatic Macrophage as a Key Player in Fatty Liver Disease. Front Immunol 2021; 12:708978. [PMID: 34956171 PMCID: PMC8696173 DOI: 10.3389/fimmu.2021.708978] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2021] [Accepted: 11/16/2021] [Indexed: 12/13/2022] Open
Abstract
Fatty liver disease, characterized by excessive inflammation and lipid deposition, is becoming one of the most prevalent liver metabolic diseases worldwide owing to the increasing global incidence of obesity. However, the underlying mechanisms of fatty liver disease are poorly understood. Accumulating evidence suggests that hepatic macrophages, specifically Kupffer cells (KCs), act as key players in the progression of fatty liver disease. Thus, it is essential to examine the current evidence of the roles of hepatic macrophages (both KCs and monocyte-derived macrophages). In this review, we primarily address the heterogeneities and multiple patterns of hepatic macrophages participating in the pathogenesis of fatty liver disease, including Toll-like receptors (TLRs), NLRP3 inflammasome, lipotoxicity, glucotoxicity, metabolic reprogramming, interaction with surrounding cells in the liver, and iron poisoning. A better understanding of the diverse roles of hepatic macrophages in the development of fatty liver disease may provide a more specific and promising macrophage-targeting therapeutic strategy for inflammatory liver diseases.
Collapse
Affiliation(s)
- Liyun Xu
- Key Laboratory for Experimental Teratology of Ministry of Education, Shandong Key Laboratory of Infection and Immunity and Department of Immunology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China.,Cell and Molecular Biology Laboratory, Zhoushan Hospital, Zhoushan, China
| | - Wen Liu
- Key Laboratory for Experimental Teratology of Ministry of Education, Shandong Key Laboratory of Infection and Immunity and Department of Immunology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China.,Institute of Basic Medicine Sciences, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Fuxiang Bai
- Laboratory for Tissue Engineering and Regeneration, School of Stomatology, Shandong University, Jinan, China
| | - Yong Xu
- Key Laboratory for Experimental Teratology of Ministry of Education, Shandong Key Laboratory of Infection and Immunity and Department of Immunology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China.,Department of Laboratory, Yueyang Hospital, Hunan Normal University, Yueyang, China
| | - Xiaohong Liang
- Key Laboratory for Experimental Teratology of Ministry of Education, Shandong Key Laboratory of Infection and Immunity and Department of Immunology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Chunhong Ma
- Key Laboratory for Experimental Teratology of Ministry of Education, Shandong Key Laboratory of Infection and Immunity and Department of Immunology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Lifen Gao
- Key Laboratory for Experimental Teratology of Ministry of Education, Shandong Key Laboratory of Infection and Immunity and Department of Immunology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, China
| |
Collapse
|
119
|
High-Fat Diet-Induced Fatty Liver Is Associated with Immunosuppressive Response during Sepsis in Mice. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:5833857. [PMID: 34925696 PMCID: PMC8674062 DOI: 10.1155/2021/5833857] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Accepted: 11/09/2021] [Indexed: 11/17/2022]
Abstract
High-fat diet-induced fatty liver is an indolent and chronic disease accompanied by immune dysfunction and metabolic disturbances involving numerous biological pathways. This study investigated how this abnormal metabolic disorder influences sepsis in mice. Mice were fed with normal chow (NC) or high-fat diet (HFD), and palmitic acid (PA) was used to treat hepatocytes to mimic fat accumulation in vitro. Lipopolysaccharide (LPS) was used to induce sepsis and related immune responses. Mice fed on a high-fat diet displayed higher mortality and more severe liver damage but compromised immunoreaction. The supernatant from PA-treated primary hepatocytes markedly diminished the inflammatory cytokine expression of macrophages after LPS stimulation, which showed a state of immunosuppression. Metabolomics analysis indicated the level of many key metabolites with possible roles in immunoreaction was altered in the HFD and PA groups compared with corresponding controls; specifically, β-hydroxybutyric acid (BHB) showed an immunosuppressive effect on Raw264.7 cells during the LPS stimulation. Transcriptomic analysis suggested that several differential signaling pathways may be associated with the alteration of immune function between the NC and HFD groups, as well as in the in vitro model. Our study suggests that the consumption of HFD may alter the hepatic metabolic profile, and that certain metabolites may remold the immune system to immunosuppressive state in the context of sepsis.
Collapse
|
120
|
Li W, Li F, Zhang X, Lin HK, Xu C. Insights into the post-translational modification and its emerging role in shaping the tumor microenvironment. Signal Transduct Target Ther 2021; 6:422. [PMID: 34924561 PMCID: PMC8685280 DOI: 10.1038/s41392-021-00825-8] [Citation(s) in RCA: 114] [Impact Index Per Article: 28.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Revised: 11/02/2021] [Accepted: 11/05/2021] [Indexed: 12/11/2022] Open
Abstract
More and more in-depth studies have revealed that the occurrence and development of tumors depend on gene mutation and tumor heterogeneity. The most important manifestation of tumor heterogeneity is the dynamic change of tumor microenvironment (TME) heterogeneity. This depends not only on the tumor cells themselves in the microenvironment where the infiltrating immune cells and matrix together forming an antitumor and/or pro-tumor network. TME has resulted in novel therapeutic interventions as a place beyond tumor beds. The malignant cancer cells, tumor infiltrate immune cells, angiogenic vascular cells, lymphatic endothelial cells, cancer-associated fibroblastic cells, and the released factors including intracellular metabolites, hormonal signals and inflammatory mediators all contribute actively to cancer progression. Protein post-translational modification (PTM) is often regarded as a degradative mechanism in protein destruction or turnover to maintain physiological homeostasis. Advances in quantitative transcriptomics, proteomics, and nuclease-based gene editing are now paving the global ways for exploring PTMs. In this review, we focus on recent developments in the PTM area and speculate on their importance as a critical functional readout for the regulation of TME. A wealth of information has been emerging to prove useful in the search for conventional therapies and the development of global therapeutic strategies.
Collapse
Affiliation(s)
- Wen Li
- Integrative Cancer Center & Cancer Clinical Research Center, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, School of Medicine, University of Electronic Science and Technology of China, 610042, Chengdu, P. R. China
| | - Feifei Li
- Integrative Cancer Center & Cancer Clinical Research Center, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, School of Medicine, University of Electronic Science and Technology of China, 610042, Chengdu, P. R. China
- Guangxi Collaborative Innovation Center for Biomedicine (Guangxi-ASEAN Collaborative Innovation Center for Major Disease Prevention and Treatment), Guangxi Medical University, 530021, Nanning, Guangxi, China
| | - Xia Zhang
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University (Army Medical University), 400038, Chongqing, China
| | - Hui-Kuan Lin
- Department of Cancer Biology, Wake Forest Baptist Medical Center, Wake Forest University, Winston Salem, NC, 27101, USA
| | - Chuan Xu
- Integrative Cancer Center & Cancer Clinical Research Center, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, School of Medicine, University of Electronic Science and Technology of China, 610042, Chengdu, P. R. China.
- Department of Cancer Biology, Wake Forest Baptist Medical Center, Wake Forest University, Winston Salem, NC, 27101, USA.
| |
Collapse
|
121
|
Cui C, Chakraborty K, Tang XA, Schoenfelt KQ, Hoffman A, Blank A, McBeth B, Pulliam N, Reardon CA, Kulkarni SA, Vaisar T, Ballabio A, Krishnan Y, Becker L. A lysosome-targeted DNA nanodevice selectively targets macrophages to attenuate tumours. NATURE NANOTECHNOLOGY 2021; 16:1394-1402. [PMID: 34764452 DOI: 10.1038/s41565-021-00988-z] [Citation(s) in RCA: 56] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Accepted: 08/31/2021] [Indexed: 06/13/2023]
Abstract
Activating CD8+ T cells by antigen cross-presentation is remarkably effective at eliminating tumours. Although this function is traditionally attributed to dendritic cells, tumour-associated macrophages (TAMs) can also cross-present antigens. TAMs are the most abundant tumour-infiltrating leukocyte. Yet, TAMs have not been leveraged to activate CD8+ T cells because mechanisms that modulate their ability to cross-present antigens are incompletely understood. Here we show that TAMs harbour hyperactive cysteine protease activity in their lysosomes, which impedes antigen cross-presentation, thereby preventing CD8+ T cell activation. We developed a DNA nanodevice (E64-DNA) that targets the lysosomes of TAMs in mice. E64-DNA inhibits the population of cysteine proteases that is present specifically inside the lysosomes of TAMs, improves their ability to cross-present antigens and attenuates tumour growth via CD8+ T cells. When combined with cyclophosphamide, E64-DNA showed sustained tumour regression in a triple-negative-breast-cancer model. Our studies demonstrate that DNA nanodevices can be targeted with organelle-level precision to reprogram macrophages and achieve immunomodulation in vivo.
Collapse
Affiliation(s)
- Chang Cui
- Committee on Cancer Biology, The University of Chicago, Chicago, IL, USA
- Ben May Department for Cancer Research, The University of Chicago, Chicago, IL, USA
| | - Kasturi Chakraborty
- Ben May Department for Cancer Research, The University of Chicago, Chicago, IL, USA
- Department of Chemistry, The University of Chicago, Chicago, IL, USA
| | - Xu Anna Tang
- Ben May Department for Cancer Research, The University of Chicago, Chicago, IL, USA
| | - Kelly Q Schoenfelt
- Ben May Department for Cancer Research, The University of Chicago, Chicago, IL, USA
| | - Alexandria Hoffman
- Ben May Department for Cancer Research, The University of Chicago, Chicago, IL, USA
| | - Ariane Blank
- Ben May Department for Cancer Research, The University of Chicago, Chicago, IL, USA
| | - Blake McBeth
- Ben May Department for Cancer Research, The University of Chicago, Chicago, IL, USA
| | - Natalie Pulliam
- Department of Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Catherine A Reardon
- Ben May Department for Cancer Research, The University of Chicago, Chicago, IL, USA
| | - Swati A Kulkarni
- Department of Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Tomas Vaisar
- UW Medicine Diabetes Institute, University of Washington, Seattle, WA, USA
| | - Andrea Ballabio
- Telethon Institute of Genetics and Medicine (TIGEM), Naples, Italy
- Department of Translational Medicine, Federico II University, Naples, Italy
- Neurological Research Institute, Baylor College of Medicine, Houston, TX, USA
| | - Yamuna Krishnan
- Department of Chemistry, The University of Chicago, Chicago, IL, USA.
- Grossman Institute of Neuroscience, Quantitative Biology and Human Behavior, The University of Chicago, Chicago, IL, USA.
| | - Lev Becker
- Committee on Cancer Biology, The University of Chicago, Chicago, IL, USA.
- Ben May Department for Cancer Research, The University of Chicago, Chicago, IL, USA.
- Ludwig Center for Metastasis Research, The University of Chicago, Chicago, IL, USA.
- University of Chicago Comprehensive Cancer Center, The University of Chicago, Chicago, IL, USA.
| |
Collapse
|
122
|
Kotlyarov S, Kotlyarova A. Anti-Inflammatory Function of Fatty Acids and Involvement of Their Metabolites in the Resolution of Inflammation in Chronic Obstructive Pulmonary Disease. Int J Mol Sci 2021; 22:12803. [PMID: 34884621 PMCID: PMC8657960 DOI: 10.3390/ijms222312803] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Revised: 11/24/2021] [Accepted: 11/24/2021] [Indexed: 12/13/2022] Open
Abstract
Lipid metabolism plays an important role in many lung functions. Disorders of lipid metabolism are part of the pathogenesis of chronic obstructive pulmonary disease (COPD). Lipids are involved in numerous cross-linkages with inflammation. Recent studies strongly support the involvement of fatty acids as participants in inflammation. They are involved in the initiation and resolution of inflammation, including acting as a substrate for the formation of lipid mediators of inflammation resolution. Specialized pro-inflammatory mediators (SPMs) belonging to the classes of lipoxins, resolvins, maresins, and protectins, which are formed enzymatically from unsaturated fatty acids, are now described. Disorders of their production and function are part of the pathogenesis of COPD. SPMs are currently the subject of active research in order to find new drugs. Short-chain fatty acids are another important participant in metabolic and immune processes, and their role in the pathogenesis of COPD is of great clinical interest.
Collapse
Affiliation(s)
- Stanislav Kotlyarov
- Department of Nursing, Ryazan State Medical University, 390026 Ryazan, Russia
| | - Anna Kotlyarova
- Department of Pharmacology and Pharmacy, Ryazan State Medical University, 390026 Ryazan, Russia;
| |
Collapse
|
123
|
Ma J, Hu W, Zhang D, Xie J, Duan C, Liu Y, Wang Y, Xu X, Cheng K, Jin B, Zhang Y, Zhuang R. CD226 knockout alleviates high-fat diet induced obesity by suppressing proinflammatory macrophage phenotype. J Transl Med 2021; 19:477. [PMID: 34823548 PMCID: PMC8620575 DOI: 10.1186/s12967-021-03150-4] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Accepted: 11/17/2021] [Indexed: 12/30/2022] Open
Abstract
Obesity is associated with chronic low-grade inflammation, contributing to an increasing prevalence of chronic metabolic diseases, such as insulin resistance, non-alcoholic fatty liver disease (NALFD), and steatohepatitis. Macrophages are the predominant immune cells in adipose tissues. Adipose tissue macrophages (ATMs) would switch to pro-inflammatory M1 state during obesity, causing local and systemic inflammation. However, the regulatory mechanism of ATMs has not yet been well described within this process. Using a high-fat diet (HFD)–induced mouse obesity model, we found that the costimulatory molecule CD226 was highly expressed on ATMs and knockout (KO) of CD226 alleviated obesity caused by HFD. Loss of CD226 reduced the accumulation of ATMs and hindered macrophage M1 polarization, with lower serum proinflammatory cytokine levels. Furthermore, deficiency of CD226 on ATMs decreased the phosphorylation levels of VAV1, AKT, and FOXO1 and thereby upregulated PPAR-γ. Further administration of PPAR-γ inhibitor restored M1 phenotype in CD226KO ATMs. In summary, loss of CD226 alleviates the HFD-induced obesity and systemic inflammation through inhibition of the accumulation and M1 polarization of ATMs in which PPAR-γ-dependent signaling pathway is involved, suggesting that CD226 may be identified as a potential molecular target for the clinical treatment of obesity.
Collapse
Affiliation(s)
- Jingchang Ma
- Department of Immunology, Fourth Military Medical University, 169 West Changle Road, Xi'an, 710032, Shaanxi, China
| | - Wei Hu
- Department of Immunology, Fourth Military Medical University, 169 West Changle Road, Xi'an, 710032, Shaanxi, China
| | - Dongliang Zhang
- Department of Immunology, Fourth Military Medical University, 169 West Changle Road, Xi'an, 710032, Shaanxi, China
| | - Jiangang Xie
- Department of Immunology, Fourth Military Medical University, 169 West Changle Road, Xi'an, 710032, Shaanxi, China
| | - Chujun Duan
- Department of Immunology, Fourth Military Medical University, 169 West Changle Road, Xi'an, 710032, Shaanxi, China
| | - Yitian Liu
- Department of Immunology, Fourth Military Medical University, 169 West Changle Road, Xi'an, 710032, Shaanxi, China
| | - Yuling Wang
- Department of Immunology, Fourth Military Medical University, 169 West Changle Road, Xi'an, 710032, Shaanxi, China
| | - Xuexue Xu
- Institute of Medical Research, Northwestern Polytechnical University, 127 West Youyi Road, Xi'an, 710072, Shaanxi, China
| | - Kun Cheng
- Department of Immunology, Fourth Military Medical University, 169 West Changle Road, Xi'an, 710032, Shaanxi, China
| | - Boquan Jin
- Department of Immunology, Fourth Military Medical University, 169 West Changle Road, Xi'an, 710032, Shaanxi, China
| | - Yuan Zhang
- Institute of Medical Research, Northwestern Polytechnical University, 127 West Youyi Road, Xi'an, 710072, Shaanxi, China.
| | - Ran Zhuang
- Department of Immunology, Fourth Military Medical University, 169 West Changle Road, Xi'an, 710032, Shaanxi, China. .,Institute of Medical Research, Northwestern Polytechnical University, 127 West Youyi Road, Xi'an, 710072, Shaanxi, China.
| |
Collapse
|
124
|
Yang Q, Zhang H, Wei T, Lin A, Sun Y, Luo P, Zhang J. Single-Cell RNA Sequencing Reveals the Heterogeneity of Tumor-Associated Macrophage in Non-Small Cell Lung Cancer and Differences Between Sexes. Front Immunol 2021; 12:756722. [PMID: 34804043 PMCID: PMC8602907 DOI: 10.3389/fimmu.2021.756722] [Citation(s) in RCA: 64] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Accepted: 10/21/2021] [Indexed: 12/25/2022] Open
Abstract
Non-Small Cell Lung Cancer (NSCLC) is a disease with high morbidity and mortality, which has sex-related differences in prognosis and immunotherapy efficacy. However, the difference in the mechanisms remains unclear. Macrophages, characterized by high plasticity and heterogeneity, act as one of the key cells that exert anti-tumor effects in the tumor microenvironment (TME) and play a complicated role in the process of tumor progression. To elucidate the subtype composition and functional heterogeneity of tumor-associated macrophages (TAMs) in NSCLC and further compare the sex-mediated differences, we conducted a single-cell level analysis in early-stage smoking NSCLC patients, combined with ssGSEA analysis, pseudotime ordering, and SCENIC analysis. We found two universally presented immune-suppressive TAMs with different functional and metabolic characteristics in the TME of NSCLC. Specifically, CCL18+ macrophages exerted immune-suppressive effects by inhibiting the production of inflammatory factors and manifested high levels of fatty acid oxidative phosphorylation metabolism. Conversely, the main metabolism pathway for SPP1+ macrophage was glycolysis which contributed to tumor metastasis by promoting angiogenesis and matrix remodeling. In terms of the differentially expressed genes, the complement gene C1QC and the matrix remodeling relevant genes FN1 and SPP1 were differentially expressed in the TAMs between sexes, of which the male upregulated SPP1 showed the potential as an ideal target for adjuvant immunotherapy and improving the efficacy of immunotherapy. According to the early-stage TCGA-NSCLC cohort, high expression of the above three genes in immune cells were associated with poor prognosis and acted as independent prognostic factors. Moreover, through verification at the transcription factor, transcriptome, and protein levels, we found that TAMs from women showed stronger immunogenicity with higher interferon-producing and antigen-presenting ability, while men-derived TAMs upregulated the PPARs and matrix remodeling related pathways, thus were more inclined to be immunosuppressive. Deconstruction of the TAMs at the single-cell level deepens our understanding of the mechanism for tumor occurrence and progress, which could be helpful to achieve the precise sex-specific tumor treatment sooner.
Collapse
Affiliation(s)
- Qi Yang
- Department of Oncology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Hongman Zhang
- Department of Oncology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Ting Wei
- Department of Oncology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Anqi Lin
- Department of Oncology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Yueqin Sun
- Department of Oncology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Peng Luo
- Department of Oncology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Jian Zhang
- Department of Oncology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| |
Collapse
|
125
|
Qin Y, Li B, Arumugam S, Lu Q, Mankash SM, Li J, Sun B, Li J, Flavell RA, Li HB, Ouyang X. m 6A mRNA methylation-directed myeloid cell activation controls progression of NAFLD and obesity. Cell Rep 2021; 37:109968. [PMID: 34758326 PMCID: PMC8667589 DOI: 10.1016/j.celrep.2021.109968] [Citation(s) in RCA: 74] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Revised: 08/02/2021] [Accepted: 10/19/2021] [Indexed: 12/23/2022] Open
Abstract
N6-methyladenosine (m6A) RNA modification is a fundamental determinant of mRNA metabolism, but its role in innate immunity-driven non-alcoholic fatty liver disease (NAFLD) and obesity is not known. Here, we show that myeloid lineage-restricted deletion of the m6A "writer" protein Methyltransferase Like 3 (METTL3) prevents age-related and diet-induced development of NAFLD and obesity in mice with improved inflammatory and metabolic phenotypes. Mechanistically, loss of METTL3 results in the differential expression of multiple mRNA transcripts marked with m6A, with a notable increase of DNA Damage Inducible Transcript 4 (DDIT4) mRNA level. In METTL3-deficient macrophages, there is a significant downregulation of mammalian target of rapamycin (mTOR) and nuclear factor κB (NF-κB) pathway activity in response to cellular stress and cytokine stimulation, which can be restored by knockdown of DDIT4. Taken together, our findings identify the contribution of METTL3-mediated m6A modification of Ddit4 mRNA to macrophage metabolic reprogramming in NAFLD and obesity.
Collapse
Affiliation(s)
- Yanqin Qin
- Department of Internal Medicine, Section of Digestive Diseases, Yale University School of Medicine, New Haven, CT 06520, USA; Henan Key Laboratory of Chinese Medicine for Respiratory Disease, Co-construction Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases by Henan & Education Ministry of P.R. China, Henan University of Chinese Medicine, Zhengzhou, Henan 450046, China
| | - Binghua Li
- Department of Internal Medicine, Section of Digestive Diseases, Yale University School of Medicine, New Haven, CT 06520, USA; Department of Hepatobiliary Surgery, The Affiliated Drum Tower Hospital, Nanjing University Medical School, Nanjing 210008, China
| | - Suyavaran Arumugam
- Department of Internal Medicine, Section of Digestive Diseases, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Qiuxia Lu
- Department of Internal Medicine, Section of Digestive Diseases, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Salah M Mankash
- Department of Internal Medicine, Section of Digestive Diseases, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Junzi Li
- Department of Internal Medicine, Section of Digestive Diseases, Yale University School of Medicine, New Haven, CT 06520, USA; Henan Key Laboratory of Chinese Medicine for Respiratory Disease, Co-construction Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases by Henan & Education Ministry of P.R. China, Henan University of Chinese Medicine, Zhengzhou, Henan 450046, China
| | - Beicheng Sun
- Department of Hepatobiliary Surgery, The Affiliated Drum Tower Hospital, Nanjing University Medical School, Nanjing 210008, China
| | - Jiansheng Li
- Henan Key Laboratory of Chinese Medicine for Respiratory Disease, Co-construction Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases by Henan & Education Ministry of P.R. China, Henan University of Chinese Medicine, Zhengzhou, Henan 450046, China
| | - Richard A Flavell
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06520, USA; Howard Hughes Medical Institute, Yale University School of Medicine, New Haven, CT 06520, USA.
| | - Hua-Bing Li
- Shanghai Institute of Immunology, State Key Laboratory of Oncogenes and Related Genes, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China; Shanghai Jiao Tong University School of Medicine-Yale Institute for Immune Metabolism, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China.
| | - Xinshou Ouyang
- Department of Internal Medicine, Section of Digestive Diseases, Yale University School of Medicine, New Haven, CT 06520, USA.
| |
Collapse
|
126
|
Oh J, Lee Y, Oh SW, Li T, Shin J, Park SH, Lee J. The Role of Adiponectin in the Skin. Biomol Ther (Seoul) 2021; 30:221-231. [PMID: 34615771 PMCID: PMC9047493 DOI: 10.4062/biomolther.2021.089] [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: 05/08/2021] [Revised: 07/29/2021] [Accepted: 08/20/2021] [Indexed: 11/29/2022] Open
Abstract
Adiponectin (Ad), a 30 kDa molecule, is an anti-diabetic adipokine; although derived from adipose tissue, it performs numerous activities in various other tissues. It binds to its own receptors, namely adiponectin receptor 1(AdipoR1), adiponectin receptor 2 (AdipoR2), and T-cadherin (CDH13). Ad plays several roles, especially as a regulator. It modulates lipid and glucose metabolism and promotes insulin sensitivity. This demonstrates that Ad has a robust correlation with fat metabolism. Furthermore, although Ad is not in direct contact with other tissues, including the skin, it can be delivered to them by diffusion or secretion via the endocrine system. Recently it has been reported that Ad can impact skin cell biology, underscoring its potential as a therapeutic biomarker of skin diseases. In the present review, we have discussed the association between skin cell biology and Ad. To elaborate further, we described the involvement of Ad in the biology of various types of cells in the skin, such as keratinocytes, fibroblasts, melanocytes, and immune cells. Additionally, we postulated that Ad could be employed as a therapeutic target to maintain skin homeostasis.
Collapse
Affiliation(s)
- Jieun Oh
- Department of Integrative Biotechnology, College of Biotechnology and Bioengineering, Sungkyunkwan University, Suwon 16419
| | - Yeongyeong Lee
- Department of Integrative Biotechnology, College of Biotechnology and Bioengineering, Sungkyunkwan University, Suwon 16419
| | - Sae-Woong Oh
- Molecular Dermatology Laboratory, Department of Integrative Biotechnology, College of Biotechnology and Bioengineering, Sungkyunkwan University, Suwon 16419
| | - TianTian Li
- Department of Integrative Biotechnology, College of Biotechnology and Bioengineering, Sungkyunkwan University, Suwon 16419
| | - Jiwon Shin
- Department of Integrative Biotechnology, College of Biotechnology and Bioengineering, Sungkyunkwan University, Suwon 16419
| | - See-Hyoung Park
- Department of Bio and Chemical Engineering, Hongik University, Sejong 30016, Republic of Korea
| | - Jongsung Lee
- Department of Integrative Biotechnology, College of Biotechnology and Bioengineering, Sungkyunkwan University, Suwon 16419.,Molecular Dermatology Laboratory, Department of Integrative Biotechnology, College of Biotechnology and Bioengineering, Sungkyunkwan University, Suwon 16419
| |
Collapse
|
127
|
Aria H, Ghaedrahmati F, Ganjalikhani-Hakemi M. Cutting edge: Metabolic immune reprogramming, reactive oxygen species, and cancer. J Cell Physiol 2021; 236:6168-6189. [PMID: 33561318 DOI: 10.1002/jcp.30303] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2020] [Revised: 01/09/2021] [Accepted: 01/19/2021] [Indexed: 02/05/2023]
Abstract
A recently proposed term "immunometabolism" points to the functional intracellular metabolic changes that occur within different immune cells. Recent findings suggest that immune responses can be determined by the metabolic status of immune cells and metabolic reprogramming is an important feature of immune cell activation. Metabolic reprogramming is also well known for cancer cells and has been suggested as a major sign of cancer progression. Metabolic reprogramming of immune cells is also seen in the tumor microenvironment. In the past decade, immunometabolism has progressively become an extraordinarily vibrant and productive area of study in immunology because of its importance for immunotherapy. Understanding the immunometabolic situation of T cells and other immune cells along with the metabolic behavior of cancer cells can help us design new therapeutic approaches against cancers. Here, we have the aim to review the cutting-edge findings on the immunometabolic situation in immune and tumor cells. We discuss new findings on signaling pathways during metabolic reprogramming, its regulation, and the participation of reactive oxygen species in these processes.
Collapse
Affiliation(s)
- Hamid Aria
- Department of Immunology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Farhoodeh Ghaedrahmati
- Department of Immunology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | | |
Collapse
|
128
|
Xia Y, Brown ZJ, Huang H, Tsung A. Metabolic reprogramming of immune cells: Shaping the tumor microenvironment in hepatocellular carcinoma. Cancer Med 2021; 10:6374-6383. [PMID: 34390203 PMCID: PMC8446566 DOI: 10.1002/cam4.4177] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Revised: 06/26/2021] [Accepted: 07/19/2021] [Indexed: 12/13/2022] Open
Abstract
Hepatocellular carcinoma (HCC) is a typical inflammation‐induced cancer and displays a complex interaction between the tumor microenvironment and tumor development. Immune cells in the HCC microenvironment play both pro‐ and anti‐tumoral roles in HCC progression. An increasing number of findings indicate that metabolic reprogramming is essential for immune cell differentiation and function. In this review, we discuss the metabolic changes of different immune cells and correlate these findings to HCC progression.
Collapse
Affiliation(s)
- Yujia Xia
- Division of Surgical Oncology, Department of Surgery, The Ohio State University Wexner Medical Center, Columbus, OH, USA.,Department of Gastroenterology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Zachary J Brown
- Division of Surgical Oncology, Department of Surgery, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Hai Huang
- Division of Surgical Oncology, Department of Surgery, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Allan Tsung
- Division of Surgical Oncology, Department of Surgery, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| |
Collapse
|
129
|
Jin R, Hao J, Yi Y, Sauter E, Li B. Regulation of macrophage functions by FABP-mediated inflammatory and metabolic pathways. Biochim Biophys Acta Mol Cell Biol Lipids 2021; 1866:158964. [PMID: 33984518 PMCID: PMC8169605 DOI: 10.1016/j.bbalip.2021.158964] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Revised: 04/26/2021] [Accepted: 04/27/2021] [Indexed: 12/15/2022]
Abstract
Macrophages are almost everywhere in the body, where they serve pivotal functions in maintaining tissue homeostasis, remodeling, and immunoregulation. Macrophages are traditionally thought to differentiate from bone marrow-derived hematopoietic stem cells (HSCs). Emerging studies suggest that some tissue macrophages at steady state originate from embryonic precursors in the yolk sac or fetal liver and are maintained in situ by self-renewal, but bone marrow-derived monocytes can give rise to tissue macrophages in pathogenic settings, such as inflammatory injuries and cancer. Macrophages are popularly classified as Th1 cytokine (e.g. IFNγ)-activated M1 macrophages (the classical activation) or Th2 cytokine (e.g. IL-4)-activated M2 macrophages (the alternative activation). However, given the myriad arrays of stimuli macrophages may encounter from local environment, macrophages exhibit notorious heterogeneity in their phenotypes and functions. Determining the underlying metabolic pathways engaged during macrophage activation is critical for understanding macrophage phenotypic and functional adaptivity under different disease settings. Fatty acid binding proteins (FABPs) represent a family of evolutionarily conserved proteins facilitating lipid transport, metabolism and responses inside cells. More specifically, adipose-FABP (A-FABP) and epidermal-FABP (E-FABP) are highly expressed in macrophages and play a central role in integrating metabolic and inflammatory pathways. In this review we highlight how A-FABP and E-FABP are respectively upregulated in different subsets of activated macrophages and provide a unique perspective in defining macrophage phenotypic and functional heterogeneity through FABP-regulated lipid metabolic and inflammatory pathways.
Collapse
Affiliation(s)
- Rong Jin
- Department of Microbiology and Immunology, University of Louisville, Louisville, KY, USA; Department of Immunology, School of Basic Medical Sciences, Peking University, Beijing, China
| | - Jiaqing Hao
- Department of Microbiology and Immunology, University of Louisville, Louisville, KY, USA
| | - Yanmei Yi
- Department of Microbiology and Immunology, University of Louisville, Louisville, KY, USA; School of Basic Medical Sciences, Guangdong Medical University, Zhanjiang, China
| | - Edward Sauter
- Division of Cancer Prevention, NIH/NCI, Bethesda, MD, USA
| | - Bing Li
- Department of Microbiology and Immunology, University of Louisville, Louisville, KY, USA.
| |
Collapse
|
130
|
Königshofer P, Brusilovskaya K, Petrenko O, Hofer BS, Schwabl P, Trauner M, Reiberger T. Nuclear Receptors in Liver Fibrosis. Biochim Biophys Acta Mol Basis Dis 2021; 1867:166235. [PMID: 34339839 DOI: 10.1016/j.bbadis.2021.166235] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Revised: 07/18/2021] [Accepted: 07/27/2021] [Indexed: 12/12/2022]
Abstract
Nuclear receptors are ligand-activated transcription factors that regulate gene expression of a variety of key molecular signals involved in liver fibrosis. The primary cellular driver of liver fibrogenesis are activated hepatic stellate cells. Different NRs regulate the hepatic expression of pro-inflammatory and pro-fibrogenic cytokines that promote the transformation of hepatic stellate cells into fibrogenic myofibroblasts. Importantly, nuclear receptors regulate gene expression circuits that promote hepatic fibrogenesis and/or allow liver fibrosis regression. In this review, we highlight the direct and indirect influence of nuclear receptors on liver fibrosis, with a focus on hepatic stellate cells, and discuss potential therapeutic effects of nuclear receptor modulation in regard to anti-fibrotic and anti-inflammatory effects. Further research on nuclear receptors-related signaling may lead to the clinical development of effective anti-fibrotic therapies for patients with liver disease.
Collapse
Affiliation(s)
- Philipp Königshofer
- Division of Gastroenterology and Hepatology, Department of Medicine III, Medical University of Vienna, Vienna, Austria; Vienna Experimental Hepatic Hemodynamic Lab (HEPEX), Medical University of Vienna, Vienna, Austria; Christian Doppler Lab for Portal Hypertension and Liver Fibrosis, Medical University of Vienna, Vienna, Austria
| | - Ksenia Brusilovskaya
- Division of Gastroenterology and Hepatology, Department of Medicine III, Medical University of Vienna, Vienna, Austria; Vienna Experimental Hepatic Hemodynamic Lab (HEPEX), Medical University of Vienna, Vienna, Austria; Christian Doppler Lab for Portal Hypertension and Liver Fibrosis, Medical University of Vienna, Vienna, Austria
| | - Oleksandr Petrenko
- Division of Gastroenterology and Hepatology, Department of Medicine III, Medical University of Vienna, Vienna, Austria; Vienna Experimental Hepatic Hemodynamic Lab (HEPEX), Medical University of Vienna, Vienna, Austria; Christian Doppler Lab for Portal Hypertension and Liver Fibrosis, Medical University of Vienna, Vienna, Austria; Ludwig Boltzmann Institute for Rare and Undiagnosed Diseases, Vienna, Austria; CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Benedikt Silvester Hofer
- Division of Gastroenterology and Hepatology, Department of Medicine III, Medical University of Vienna, Vienna, Austria; Vienna Experimental Hepatic Hemodynamic Lab (HEPEX), Medical University of Vienna, Vienna, Austria; Christian Doppler Lab for Portal Hypertension and Liver Fibrosis, Medical University of Vienna, Vienna, Austria
| | - Philipp Schwabl
- Division of Gastroenterology and Hepatology, Department of Medicine III, Medical University of Vienna, Vienna, Austria; Vienna Experimental Hepatic Hemodynamic Lab (HEPEX), Medical University of Vienna, Vienna, Austria; Christian Doppler Lab for Portal Hypertension and Liver Fibrosis, Medical University of Vienna, Vienna, Austria
| | - Michael Trauner
- Division of Gastroenterology and Hepatology, Department of Medicine III, Medical University of Vienna, Vienna, Austria
| | - Thomas Reiberger
- Division of Gastroenterology and Hepatology, Department of Medicine III, Medical University of Vienna, Vienna, Austria; Vienna Experimental Hepatic Hemodynamic Lab (HEPEX), Medical University of Vienna, Vienna, Austria; Christian Doppler Lab for Portal Hypertension and Liver Fibrosis, Medical University of Vienna, Vienna, Austria; Ludwig Boltzmann Institute for Rare and Undiagnosed Diseases, Vienna, Austria; CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria.
| |
Collapse
|
131
|
Wu H, Wang M, Li X, Shao Y. The Metaflammatory and Immunometabolic Role of Macrophages and Microglia in Diabetic Retinopathy. Hum Cell 2021; 34:1617-1628. [PMID: 34324139 DOI: 10.1007/s13577-021-00580-6] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Accepted: 07/17/2021] [Indexed: 12/17/2022]
Abstract
Emergent studies reveal the roles of inflammatory cells and cytokines in the development of diabetic retinopathy (DR), which is gradually portrayed as a chronic inflammatory disease accompanied by metabolic disorder. Through the pathogenesis of DR, macrophages or microglia play a critical role in the inflammation, neovascularization, and neurodegeneration of the retina. Conventionally, macrophages are generally divided into M1 and M2 phenotypes which mainly rely on glycolysis and oxidative phosphorylation, respectively. Recently, studies have found that nutrients (including glucose and lipids) and metabolites (such as lactate), can not only provide energy for cells, but also act as signaling molecules to regulate the function and fate of cells. In this review, we discussed the intrinsic correlations among the metabolic status, polarization, and function of macrophage/microglia in DR. Hyperglycemia and hyperlipidemia could induce M1-like and M2-like macrophages polarization in different phases of DR. Targeting the regulation of microglial metabolic profile might be a promising therapeutic strategy to modulate the polarization and function of macrophages/microglia, thus attenuating the progression of DR.
Collapse
Affiliation(s)
- Honglian Wu
- Tianjin Key Laboratory of Retinal Functions and Diseases, Tianjin Medical University Eye Hospital, No. 251, Fukang Road, Nankai District, Tianjin, 300384, China.,Tianjin International Joint Research and Development Centre of Ophthalmology and Vision Science, Tianjin Medical University Eye Hospital, No. 251, Fukang Road, Nankai District, Tianjin, 300384, China.,Eye Institute and School of Optometry, Tianjin Medical University Eye Hospital, No. 251, Fukang Road, Nankai District, Tianjin, 300384, China
| | - Mengqi Wang
- Tianjin Key Laboratory of Retinal Functions and Diseases, Tianjin Medical University Eye Hospital, No. 251, Fukang Road, Nankai District, Tianjin, 300384, China.,Tianjin International Joint Research and Development Centre of Ophthalmology and Vision Science, Tianjin Medical University Eye Hospital, No. 251, Fukang Road, Nankai District, Tianjin, 300384, China.,Eye Institute and School of Optometry, Tianjin Medical University Eye Hospital, No. 251, Fukang Road, Nankai District, Tianjin, 300384, China
| | - Xiaorong Li
- Tianjin Key Laboratory of Retinal Functions and Diseases, Tianjin Medical University Eye Hospital, No. 251, Fukang Road, Nankai District, Tianjin, 300384, China.,Tianjin International Joint Research and Development Centre of Ophthalmology and Vision Science, Tianjin Medical University Eye Hospital, No. 251, Fukang Road, Nankai District, Tianjin, 300384, China.,Eye Institute and School of Optometry, Tianjin Medical University Eye Hospital, No. 251, Fukang Road, Nankai District, Tianjin, 300384, China
| | - Yan Shao
- Tianjin Key Laboratory of Retinal Functions and Diseases, Tianjin Medical University Eye Hospital, No. 251, Fukang Road, Nankai District, Tianjin, 300384, China. .,Tianjin International Joint Research and Development Centre of Ophthalmology and Vision Science, Tianjin Medical University Eye Hospital, No. 251, Fukang Road, Nankai District, Tianjin, 300384, China. .,Eye Institute and School of Optometry, Tianjin Medical University Eye Hospital, No. 251, Fukang Road, Nankai District, Tianjin, 300384, China.
| |
Collapse
|
132
|
Ye L, Huang W, Liu S, Cai S, Hong L, Xiao W, Thiele K, Zeng Y, Song M, Diao L. Impacts of Immunometabolism on Male Reproduction. Front Immunol 2021; 12:658432. [PMID: 34367130 PMCID: PMC8334851 DOI: 10.3389/fimmu.2021.658432] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Accepted: 06/29/2021] [Indexed: 12/24/2022] Open
Abstract
The physiological process of male reproduction relies on the orchestration of neuroendocrine, immune, and energy metabolism. Spermatogenesis is controlled by the hypothalamic-pituitary-testicular (HPT) axis, which modulates the production of gonadal steroid hormones in the testes. The immune cells and cytokines in testes provide a protective microenvironment for the development and maturation of germ cells. The metabolic cellular responses and processes in testes provide energy production and biosynthetic precursors to regulate germ cell development and control testicular immunity and inflammation. The metabolism of immune cells is crucial for both inflammatory and anti-inflammatory responses, which supposes to affect the spermatogenesis in testes. In this review, the role of immunometabolism in male reproduction will be highlighted. Obesity, metabolic dysfunction, such as type 2 diabetes mellitus, are well documented to impact male fertility; thus, their impacts on the immune cells distributed in testes will also be discussed. Finally, the potential significance of the medicine targeting the specific metabolic intermediates or immune metabolism checkpoints to improve male reproduction will also be reassessed.
Collapse
Affiliation(s)
- Lijun Ye
- Shenzhen Key Laboratory for Reproductive Immunology of Peri-implantation, Clinical Research Center for Reproductive Medicine, Shenzhen Zhongshan Urology Hospital, Shenzhen, China
| | - Wensi Huang
- Shenzhen Key Laboratory for Reproductive Immunology of Peri-implantation, Clinical Research Center for Reproductive Medicine, Shenzhen Zhongshan Urology Hospital, Shenzhen, China
| | - Su Liu
- Shenzhen Key Laboratory for Reproductive Immunology of Peri-implantation, Clinical Research Center for Reproductive Medicine, Shenzhen Zhongshan Urology Hospital, Shenzhen, China
| | - Songchen Cai
- Shenzhen Key Laboratory for Reproductive Immunology of Peri-implantation, Clinical Research Center for Reproductive Medicine, Shenzhen Zhongshan Urology Hospital, Shenzhen, China
| | - Ling Hong
- Shenzhen Key Laboratory for Reproductive Immunology of Peri-implantation, Clinical Research Center for Reproductive Medicine, Shenzhen Zhongshan Urology Hospital, Shenzhen, China
| | - Weiqiang Xiao
- Shenzhen Zhongshan Institute for Reproduction and Genetics, Fertility Center, Shenzhen Zhongshan Urology Hospital, Shenzhen, China
| | - Kristin Thiele
- Division of Experimental Feto-Maternal Medicine, Department of Obstetrics and Fetal Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Yong Zeng
- Shenzhen Key Laboratory for Reproductive Immunology of Peri-implantation, Clinical Research Center for Reproductive Medicine, Shenzhen Zhongshan Urology Hospital, Shenzhen, China
| | - Mingzhe Song
- Shenzhen Zhongshan Institute for Reproduction and Genetics, Fertility Center, Shenzhen Zhongshan Urology Hospital, Shenzhen, China
| | - Lianghui Diao
- Shenzhen Key Laboratory for Reproductive Immunology of Peri-implantation, Clinical Research Center for Reproductive Medicine, Shenzhen Zhongshan Urology Hospital, Shenzhen, China
| |
Collapse
|
133
|
Larson-Casey JL, Gu L, Davis D, Cai GQ, Ding Q, He C, Carter AB. Post-translational regulation of PGC-1α modulates fibrotic repair. FASEB J 2021; 35:e21675. [PMID: 34038004 PMCID: PMC8252570 DOI: 10.1096/fj.202100339r] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Revised: 04/23/2021] [Accepted: 05/04/2021] [Indexed: 12/12/2022]
Abstract
Idiopathic pulmonary fibrosis (IPF) is a progressive lung disease associated with mitochondrial oxidative stress. Mitochondrial reactive oxygen species (mtROS) are important for cell homeostasis by regulating mitochondrial dynamics. Here, we show that IPF BAL cells exhibited increased mitochondrial biogenesis that is, in part, due to increased nuclear expression of peroxisome proliferator-activated receptor-ɣ (PPARɣ) coactivator (PGC)-1α. Increased PPARGC1A mRNA expression directly correlated with reduced pulmonary function in IPF subjects. Oxidant-mediated activation of the p38 MAPK via Akt1 regulated PGC-1α activation to increase mitochondrial biogenesis in monocyte-derived macrophages. Demonstrating the importance of PGC-1α in fibrotic repair, mice harboring a conditional deletion of Ppargc1a in monocyte-derived macrophages or mice administered a chemical inhibitor of mitochondrial division had reduced biogenesis and increased apoptosis, and the mice were protected from pulmonary fibrosis. These observations suggest that Akt1-mediated regulation of PGC-1α maintains mitochondrial homeostasis in monocyte-derived macrophages to induce apoptosis resistance, which contributes to the pathogenesis of pulmonary fibrosis.
Collapse
Affiliation(s)
- Jennifer L Larson-Casey
- Department of Medicine, Division of Pulmonary, Allergy, and Critical Care Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Linlin Gu
- Department of Medicine, Division of Pulmonary, Allergy, and Critical Care Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Dana Davis
- Department of Medicine, Division of Pulmonary, Allergy, and Critical Care Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Guo-Qiang Cai
- Department of Anesthesiology and Perioperative Medicine, Division of Molecular and Translational Biomedicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Qiang Ding
- Department of Anesthesiology and Perioperative Medicine, Division of Molecular and Translational Biomedicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Chao He
- Department of Medicine, Division of Pulmonary, Allergy, and Critical Care Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - A Brent Carter
- Department of Medicine, Division of Pulmonary, Allergy, and Critical Care Medicine, University of Alabama at Birmingham, Birmingham, AL, USA.,Birmingham Veterans Administration Medical Center, Birmingham, AL, USA
| |
Collapse
|
134
|
Tuerhong A, Xu J, Shi S, Tan Z, Meng Q, Hua J, Liu J, Zhang B, Wang W, Yu X, Liang C. Overcoming chemoresistance by targeting reprogrammed metabolism: the Achilles' heel of pancreatic ductal adenocarcinoma. Cell Mol Life Sci 2021; 78:5505-5526. [PMID: 34131808 PMCID: PMC11072422 DOI: 10.1007/s00018-021-03866-y] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Revised: 05/04/2021] [Accepted: 05/27/2021] [Indexed: 02/07/2023]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is one of the leading causes of cancer-related death due to its late diagnosis that removes the opportunity for surgery and metabolic plasticity that leads to resistance to chemotherapy. Metabolic reprogramming related to glucose, lipid, and amino acid metabolism in PDAC not only enables the cancer to thrive and survive under hypovascular, nutrient-poor and hypoxic microenvironments, but also confers chemoresistance, which contributes to the poor prognosis of PDAC. In this review, we systematically elucidate the mechanism of chemotherapy resistance and the relationship of metabolic programming features with resistance to anticancer drugs in PDAC. Targeting the critical enzymes and/or transporters involved in glucose, lipid, and amino acid metabolism may be a promising approach to overcome chemoresistance in PDAC. Consequently, regulating metabolism could be used as a strategy against PDAC and could improve the prognosis of PDAC.
Collapse
Affiliation(s)
- Abudureyimu Tuerhong
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, 270 Dong'An Road, Shanghai, 200032, People's Republic of China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, People's Republic of China
- Pancreatic Cancer Institute, Fudan University, Shanghai, 200032, People's Republic of China
- Shanghai Pancreatic Cancer Institute, Shanghai, 200032, People's Republic of China
| | - Jin Xu
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, 270 Dong'An Road, Shanghai, 200032, People's Republic of China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, People's Republic of China
- Pancreatic Cancer Institute, Fudan University, Shanghai, 200032, People's Republic of China
- Shanghai Pancreatic Cancer Institute, Shanghai, 200032, People's Republic of China
| | - Si Shi
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, 270 Dong'An Road, Shanghai, 200032, People's Republic of China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, People's Republic of China
- Pancreatic Cancer Institute, Fudan University, Shanghai, 200032, People's Republic of China
- Shanghai Pancreatic Cancer Institute, Shanghai, 200032, People's Republic of China
| | - Zhen Tan
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, 270 Dong'An Road, Shanghai, 200032, People's Republic of China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, People's Republic of China
- Pancreatic Cancer Institute, Fudan University, Shanghai, 200032, People's Republic of China
- Shanghai Pancreatic Cancer Institute, Shanghai, 200032, People's Republic of China
| | - Qingcai Meng
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, 270 Dong'An Road, Shanghai, 200032, People's Republic of China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, People's Republic of China
- Pancreatic Cancer Institute, Fudan University, Shanghai, 200032, People's Republic of China
- Shanghai Pancreatic Cancer Institute, Shanghai, 200032, People's Republic of China
| | - Jie Hua
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, 270 Dong'An Road, Shanghai, 200032, People's Republic of China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, People's Republic of China
- Pancreatic Cancer Institute, Fudan University, Shanghai, 200032, People's Republic of China
- Shanghai Pancreatic Cancer Institute, Shanghai, 200032, People's Republic of China
| | - Jiang Liu
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, 270 Dong'An Road, Shanghai, 200032, People's Republic of China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, People's Republic of China
- Pancreatic Cancer Institute, Fudan University, Shanghai, 200032, People's Republic of China
- Shanghai Pancreatic Cancer Institute, Shanghai, 200032, People's Republic of China
| | - Bo Zhang
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, 270 Dong'An Road, Shanghai, 200032, People's Republic of China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, People's Republic of China
- Pancreatic Cancer Institute, Fudan University, Shanghai, 200032, People's Republic of China
- Shanghai Pancreatic Cancer Institute, Shanghai, 200032, People's Republic of China
| | - Wei Wang
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, 270 Dong'An Road, Shanghai, 200032, People's Republic of China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, People's Republic of China
- Pancreatic Cancer Institute, Fudan University, Shanghai, 200032, People's Republic of China
- Shanghai Pancreatic Cancer Institute, Shanghai, 200032, People's Republic of China
| | - Xianjun Yu
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, 270 Dong'An Road, Shanghai, 200032, People's Republic of China.
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, People's Republic of China.
- Pancreatic Cancer Institute, Fudan University, Shanghai, 200032, People's Republic of China.
- Shanghai Pancreatic Cancer Institute, Shanghai, 200032, People's Republic of China.
| | - Chen Liang
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, 270 Dong'An Road, Shanghai, 200032, People's Republic of China.
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, People's Republic of China.
- Pancreatic Cancer Institute, Fudan University, Shanghai, 200032, People's Republic of China.
- Shanghai Pancreatic Cancer Institute, Shanghai, 200032, People's Republic of China.
| |
Collapse
|
135
|
Li P, Hao Z, Wu J, Ma C, Xu Y, Li J, Lan R, Zhu B, Ren P, Fan D, Sun S. Comparative Proteomic Analysis of Polarized Human THP-1 and Mouse RAW264.7 Macrophages. Front Immunol 2021; 12:700009. [PMID: 34267761 PMCID: PMC8276023 DOI: 10.3389/fimmu.2021.700009] [Citation(s) in RCA: 83] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2021] [Accepted: 06/11/2021] [Indexed: 11/13/2022] Open
Abstract
Macrophages can be polarized into classically activated macrophages (M1) and alternatively activated macrophages (M2) in the immune system, performing pro-inflammatory and anti-inflammatory functions, respectively. Human THP-1 and mouse RAW264.7 cell line models have been widely used in various macrophage-associated studies, while the similarities and differences in protein expression profiles between the two macrophage models are still largely unclear. In this study, the protein expression profiles of M1 and M2 phenotypes from both THP-1 and RAW264.7 macrophages were systematically investigated using mass spectrometry-based proteomics. By quantitatively analyzing more than 5,000 proteins among different types of macrophages (M0, M1 and M2) from both cell lines, we identified a list of proteins that were uniquely up-regulated in each macrophage type and further confirmed 43 proteins that were commonly up-regulated in M1 macrophages of both cell lines. These results revealed considerable divergences of each polarization type between THP-1 and RAW264.7 macrophages. Moreover, the mRNA and protein expression of CMPK2, RSAD2, DDX58, and DHX58 were strongly up-regulated in M1 macrophages for both macrophage models. These data can serve as important resources for further studies of macrophage-associated diseases in experimental pathology using human and mouse cell line models.
Collapse
Affiliation(s)
- Pengfei Li
- College of Life Science, Northwest University, Xi'an, China
| | - Zhifang Hao
- College of Life Science, Northwest University, Xi'an, China
| | - Jingyu Wu
- College of Life Science, Northwest University, Xi'an, China
| | - Chen Ma
- College of Life Science, Northwest University, Xi'an, China
| | - Yintai Xu
- College of Life Science, Northwest University, Xi'an, China
| | - Jun Li
- College of Life Science, Northwest University, Xi'an, China
| | - Rongxia Lan
- College of Life Science, Northwest University, Xi'an, China
| | - Bojing Zhu
- College of Life Science, Northwest University, Xi'an, China
| | - Pengyu Ren
- Department of Neurosurgery, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Daidi Fan
- Shaanxi Key Laboratory of Degradable Biomedical Materials, School of Chemical Engineering, Northwest University, Xi'an, China
| | - Shisheng Sun
- College of Life Science, Northwest University, Xi'an, China
| |
Collapse
|
136
|
Xie Z, Cheng Y, Zhang Q, Hao H, Yin Y, Zang L, Wang X, Mu Y. Anti-obesity effect and mechanism of mesenchymal stem cells influence on obese mice. Open Life Sci 2021; 16:653-666. [PMID: 34222665 PMCID: PMC8234810 DOI: 10.1515/biol-2021-0061] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2020] [Revised: 05/18/2021] [Accepted: 05/19/2021] [Indexed: 12/12/2022] Open
Abstract
Mesenchymal stem cells (MSCs) can be obtained from almost all tissues and present promising therapeutic effects for metabolic diseases. Human adipose-derived MSCs (hASCs) have recently been widely studied due to their easy access and low immunity. Thus, we intended to figure out the effects and potential mechanism of hASCs on obesity in high-fat-diet (HFD)-induced obese mice. Following 16 weeks of being fed HFD, hASCs were intravenously injected. Two weeks later, body weight, body composition, and energy expenditure were evaluated. Additionally, the phenotypes of macrophages infiltrating adipose tissue were analyzed. The results revealed that hASCs administration significantly reduced adipose tissue weight, adipocyte size, and fat mass and exerted beneficial effects in serum lipid profile. This anti-obesity effect was mediated by the increased O2 consumption, CO2 production, and energy expenditure, which was further evidenced by the upregulation of uncoupling protein-1 (UCP-1) and metabolism-associated genes. Furthermore, hASCs infusion increased the amount of alternatively activated (M2) macrophages in adipose tissue, and the expression of pro-inflammatory cytokines-related genes was reduced. Taken together, these results indicated that hASCs suppressed obesity by increasing UCP-1 expression and enhancing energy expenditure, and this effect might be due to the increased M2 macrophages.
Collapse
Affiliation(s)
- Zongyan Xie
- Department of Clinical Pharmacology, Beijing Luhe Hospital Affiliated to Capital Medical University, 82 Xinhua South Road, Beijing 101149, People's Republic of China
| | - Yu Cheng
- Department of Endocrinology, The First Medical Center of PLA General Hospital, 28 Fuxing Road, Beijing 100853, People's Republic of China
| | - Qi Zhang
- Department of Endocrinology, Beijing Tiantan Hospital Affiliated to Capital Medical University, Beijing 100070, People's Republic of China
| | - Haojie Hao
- Department of Molecular Biology, Institute of Basic Medicine, The First Medical Center of PLA General Hospital, Beijing 100853, People's Republic of China
| | - Yaqi Yin
- Department of Endocrinology, The First Medical Center of PLA General Hospital, 28 Fuxing Road, Beijing 100853, People's Republic of China
| | - Li Zang
- Department of Endocrinology, The First Medical Center of PLA General Hospital, 28 Fuxing Road, Beijing 100853, People's Republic of China
| | - Xuhong Wang
- Department of Clinical Pharmacology, Beijing Luhe Hospital Affiliated to Capital Medical University, 82 Xinhua South Road, Beijing 101149, People's Republic of China
| | - Yiming Mu
- Department of Endocrinology, The First Medical Center of PLA General Hospital, 28 Fuxing Road, Beijing 100853, People's Republic of China
| |
Collapse
|
137
|
Bojan A, Parvu A, Zsoldos IA, Torok T, Farcas AD. Macrophage activation syndrome: A diagnostic challenge (Review). Exp Ther Med 2021; 22:904. [PMID: 34257717 PMCID: PMC8243343 DOI: 10.3892/etm.2021.10336] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Accepted: 04/26/2021] [Indexed: 12/13/2022] Open
Abstract
Macrophage activation syndrome (MAS) represents an acute and severe inflammatory syndrome, idiopathic (primary) or secondary to infections, rheumatic diseases, malignancies, or drugs. MAS is underdiagnosed, being confused with sepsis, adverse effects of anti-arthritic drugs or exacerbated symptoms of evolving rheumatologic or infectious diseases. Because of the late diagnosis, most patients do not benefit from effective therapy, leading to death. Elucidation of valid early diagnostic criteria of MAS would be a particularly important step in reducing the mortality due to this pathology. Thus, the purpose of this review based on 40 studies centered on the diagnostic criteria of MAS. We detailed the main diagnostic criteria and the few diagnostic scores or sets of criteria that have been recently published. The criteria most frequently encountered in the literature include: Fever, hepatosplenomegaly, hyperferritinemia, hepatopathy, coagulopathy, thrombocytopenia, hypertriglyceridemia, decrease in erythrocyte sedimentation rate and bone marrow hemophagocytosis. The most elaborate diagnostic score will result following an ongoing international project and consensus, the Delphi International Survey.
Collapse
Affiliation(s)
- Anca Bojan
- Hematology Department, 'Iuliu Hatieganu' University of Medicine and Pharmacy, 400124 Cluj-Napoca, Romania.,Hematology Department, 'Prof. Dr. Ioan Chiricuta' Oncological Insitute, 400124 Cluj-Napoca, Romania
| | - Andrada Parvu
- Hematology Department, 'Iuliu Hatieganu' University of Medicine and Pharmacy, 400124 Cluj-Napoca, Romania.,Hematology Department, 'Prof. Dr. Ioan Chiricuta' Oncological Insitute, 400124 Cluj-Napoca, Romania
| | - Iulia-Andrea Zsoldos
- Hematology Department, 'Prof. Dr. Ioan Chiricuta' Oncological Insitute, 400124 Cluj-Napoca, Romania
| | - Tunde Torok
- Hematology Department, 'Iuliu Hatieganu' University of Medicine and Pharmacy, 400124 Cluj-Napoca, Romania.,Hematology Department, 'Prof. Dr. Ioan Chiricuta' Oncological Insitute, 400124 Cluj-Napoca, Romania
| | - Anca Daniela Farcas
- Internal Medicine Department, 'Iuliu Hatieganu' University of Medicine and Pharmacy, 400006 Cluj-Napoca, Romania.,Cardiology Department, Emergency County Clinic Hospital, 400006 Cluj-Napoca, Romania
| |
Collapse
|
138
|
Luque-Campos N, Bustamante-Barrientos FA, Pradenas C, García C, Araya MJ, Bohaud C, Contreras-López R, Elizondo-Vega R, Djouad F, Luz-Crawford P, Vega-Letter AM. The Macrophage Response Is Driven by Mesenchymal Stem Cell-Mediated Metabolic Reprogramming. Front Immunol 2021; 12:624746. [PMID: 34149687 PMCID: PMC8213396 DOI: 10.3389/fimmu.2021.624746] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2020] [Accepted: 05/13/2021] [Indexed: 12/17/2022] Open
Abstract
Mesenchymal stem cells (MSCs) are multipotent adult stromal cells widely studied for their regenerative and immunomodulatory properties. They are capable of modulating macrophage plasticity depending on various microenvironmental signals. Current studies have shown that metabolic changes can also affect macrophage fate and function. Indeed, changes in the environment prompt phenotype change. Therefore, in this review, we will discuss how MSCs orchestrate macrophage’s metabolic plasticity and the impact on their function. An improved understanding of the crosstalk between macrophages and MSCs will improve our knowledge of MSC’s therapeutic potential in the context of inflammatory diseases, cancer, and tissue repair processes in which macrophages are pivotal.
Collapse
Affiliation(s)
- Noymar Luque-Campos
- Laboratorio de Inmunología Celular y Molecular, Facultad de Medicina, Universidad de los Andes, Santiago, Chile.,Centro de Investigación e Innovación Biomédica, Universidad de los Andes, Santiago, Chile.,Programa de Doctorado en Biomedicina, Facultad de Medicina, Universidad de los Andes, Santiago, Chile
| | - Felipe A Bustamante-Barrientos
- Laboratorio de Inmunología Celular y Molecular, Facultad de Medicina, Universidad de los Andes, Santiago, Chile.,Centro de Investigación e Innovación Biomédica, Universidad de los Andes, Santiago, Chile
| | - Carolina Pradenas
- Laboratorio de Inmunología Celular y Molecular, Facultad de Medicina, Universidad de los Andes, Santiago, Chile.,Centro de Investigación e Innovación Biomédica, Universidad de los Andes, Santiago, Chile.,Facultad de Ciencias, Universidad de Chile, Santiago, Chile
| | - Cynthia García
- Laboratorio de Inmunología Celular y Molecular, Facultad de Medicina, Universidad de los Andes, Santiago, Chile.,Centro de Investigación e Innovación Biomédica, Universidad de los Andes, Santiago, Chile.,Escuela de Biotecnología, Facultad de Ciencias, Universidad Mayor, Santiago, Chile
| | - María Jesús Araya
- Laboratorio de Inmunología Celular y Molecular, Facultad de Medicina, Universidad de los Andes, Santiago, Chile.,Centro de Investigación e Innovación Biomédica, Universidad de los Andes, Santiago, Chile.,Escuela de Biotecnología, Facultad de Ciencias, Universidad Mayor, Santiago, Chile
| | | | | | - Roberto Elizondo-Vega
- Laboratorio de Biología Celular, Departamento de Biología Celular, Facultad de Ciencias Biológicas, Universidad de Concepción, Concepción, Chile
| | | | - Patricia Luz-Crawford
- Laboratorio de Inmunología Celular y Molecular, Facultad de Medicina, Universidad de los Andes, Santiago, Chile.,Centro de Investigación e Innovación Biomédica, Universidad de los Andes, Santiago, Chile
| | - Ana María Vega-Letter
- Centro de Investigación e Innovación Biomédica, Universidad de los Andes, Santiago, Chile.,Cells for Cells, Regenero, Las Condes, Santiago, Chile.,Laboratory of Nano-Regenerative Medicine, Facultad de Medicina, Universidad de los Andes, Santiago, Chile
| |
Collapse
|
139
|
Xu M, Wang X, Li Y, Geng X, Jia X, Zhang L, Yang H. Arachidonic Acid Metabolism Controls Macrophage Alternative Activation Through Regulating Oxidative Phosphorylation in PPARγ Dependent Manner. Front Immunol 2021; 12:618501. [PMID: 34149684 PMCID: PMC8211451 DOI: 10.3389/fimmu.2021.618501] [Citation(s) in RCA: 54] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2020] [Accepted: 05/12/2021] [Indexed: 12/11/2022] Open
Abstract
Macrophage polarization is mainly steered by metabolic reprogramming in the tissue microenvironment, thus leading to distinct outcomes of various diseases. However, the role of lipid metabolism in the regulation of macrophage alternative activation is incompletely understood. Using human THP-1 and mouse bone marrow derived macrophage polarization models, we revealed a pivotal role for arachidonic acid metabolism in determining the phenotype of M2 macrophages. We demonstrated that macrophage M2 polarization was inhibited by arachidonic acid, but inversely facilitated by its derived metabolite prostaglandin E2 (PGE2). Furthermore, PPARγ bridges these two seemingly unrelated processes via modulating oxidative phosphorylation (OXPHOS). Through inhibiting PPARγ, PGE2 enhanced OXPHOS, resulting in the alternative activation of macrophages, which was counterweighted by the activation of PPARγ. This connection between PGE2 biosynthesis and macrophage M2 polarization also existed in human and mouse esophageal squamous cell carcinoma. Our results highlight the critical role of arachidonic acid and metabolic PGE2 as immune regulators in modulating tissue homeostasis and pathological process.
Collapse
Affiliation(s)
- Miao Xu
- West China School of Public Health/West China Fourth Hospital and Healthy Food Evaluation Research Center, Sichuan University, Chengdu, China
| | - Xiaohong Wang
- NHC Key Laboratory of Food Safety Risk Assessment, China National Center for Food Safety Risk Assessment, Beijing, China
| | - Yongning Li
- NHC Key Laboratory of Food Safety Risk Assessment, China National Center for Food Safety Risk Assessment, Beijing, China
| | - Xue Geng
- NHC Key Laboratory of Food Safety Risk Assessment, China National Center for Food Safety Risk Assessment, Beijing, China
| | - Xudong Jia
- NHC Key Laboratory of Food Safety Risk Assessment, China National Center for Food Safety Risk Assessment, Beijing, China
| | - Lishi Zhang
- West China School of Public Health/West China Fourth Hospital and Healthy Food Evaluation Research Center, Sichuan University, Chengdu, China
| | - Hui Yang
- NHC Key Laboratory of Food Safety Risk Assessment, China National Center for Food Safety Risk Assessment, Beijing, China
| |
Collapse
|
140
|
Chen JY, Zhou JK, Pan W. Immunometabolism: Towards a Better Understanding the Mechanism of Parasitic Infection and Immunity. Front Immunol 2021; 12:661241. [PMID: 34122419 PMCID: PMC8191844 DOI: 10.3389/fimmu.2021.661241] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2021] [Accepted: 05/13/2021] [Indexed: 12/26/2022] Open
Abstract
As a relatively successful pathogen, several parasites can establish long-term infection in host. This “harmonious symbiosis” status relies on the “precise” manipulation of host immunity and metabolism, however, the underlying mechanism is still largely elusive. Immunometabolism is an emerging crossed subject in recent years. It mainly discusses the regulatory mechanism of metabolic changes on reprogramming the key transcriptional and post-transcriptional events related to immune cell activation and effect, which provides a novel insight for understanding how parasites regulate the infection and immunity in hosts. The present study reviewed the current research progress on metabolic reprogramming mechanism exploited by parasites to modulate the function in various immune cells, highlighting the future exploitation of key metabolites or metabolic events to clarify the underlying mechanism of anti-parasite immunity and design novel intervention strategies against parasitic infection.
Collapse
Affiliation(s)
- Jing-Yue Chen
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogen Biology and Immunology, Xuzhou Medical University, Xuzhou, China.,The First Clinical Medicine, Xuzhou Medical University, Xuzhou, China
| | - Ji-Kai Zhou
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogen Biology and Immunology, Xuzhou Medical University, Xuzhou, China.,The First Clinical Medicine, Xuzhou Medical University, Xuzhou, China
| | - Wei Pan
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogen Biology and Immunology, Xuzhou Medical University, Xuzhou, China
| |
Collapse
|
141
|
Gao YR, Zhang RH, Li R, Tang CL, Pan Q, Pen P. The effects of helminth infections against type 2 diabetes. Parasitol Res 2021; 120:1935-1942. [PMID: 34002262 DOI: 10.1007/s00436-021-07189-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Accepted: 05/07/2021] [Indexed: 12/15/2022]
Abstract
Type 2 diabetes mellitus (T2D) is a prevalent inflammation-related disease characterized by insulin resistance and elevated blood glucose levels. The high incidence rate of T2D in Western societies may be due to environmental conditions, including reduced worm exposure. In human and animal models, some helminths, such as Schistosoma, Nippostrongylus, Strongyloides, and Heligmosomoides, and their products reportedly ameliorate or prevent T2D progression. T2D induces adaptive immune pathways involved in the inhibition of type 1 immune responses, promotion of type 2 immune responses, and expansion of regulatory T cells and innate immune cells, such as macrophages, eosinophils, and group 2 innate lymphoid cells. Among immune cells expanded in T2DM, type 2 immune cells and macrophages are the most important and may have synergistic effects. The stimulation of host immunity by helminth infections also promotes interactions between the innate and adaptive immune systems. In this paper, we provide a comprehensive review of intestinal helminths' protective effects against T2D.
Collapse
Affiliation(s)
- Yan-Ru Gao
- Medical Department, City College, Wuhan University of Science and Technology, Wuhan, 430083, China
| | - Rong-Hui Zhang
- Wuchang Hospital Affiliated to Wuhan University of Science and Technology, Wuhan, 430063, China
| | - Ru Li
- Wuchang Hospital Affiliated to Wuhan University of Science and Technology, Wuhan, 430063, China
| | - Chun-Lian Tang
- Wuchang Hospital Affiliated to Wuhan University of Science and Technology, Wuhan, 430063, China.
| | - Qun Pan
- Wuchang Hospital Affiliated to Wuhan University of Science and Technology, Wuhan, 430063, China.
| | - Peng Pen
- Wuhan Institute for Tuberculosis Control, Wuhan Pulmonary Hospital, Wuhan, 430030, China.
| |
Collapse
|
142
|
Candida Administration in Bilateral Nephrectomy Mice Elevates Serum (1→3)-β-D-glucan That Enhances Systemic Inflammation Through Energy Augmentation in Macrophages. Int J Mol Sci 2021; 22:ijms22095031. [PMID: 34068595 PMCID: PMC8126065 DOI: 10.3390/ijms22095031] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Revised: 04/18/2021] [Accepted: 04/29/2021] [Indexed: 12/15/2022] Open
Abstract
Systemic inflammation, from gut translocation of organismal molecules, might worsen uremic complications in acute kidney injury (AKI). The monitoring of gut permeability integrity and/or organismal molecules in AKI might be clinically beneficial. Due to the less prominence of Candida albicans in human intestine compared with mouse gut, C. albicans were orally administered in bilateral nephrectomy (BiN) mice. Gut dysbiosis, using microbiome analysis, and gut permeability defect (gut leakage), which was determined by fluorescein isothiocyanate-dextran and intestinal tight-junction immunofluorescent staining, in mice with BiN-Candida was more severe than BiN without Candida. Additionally, profound gut leakage in BiN-Candida also resulted in gut translocation of lipopolysaccharide (LPS) and (1→3)-β-D-glucan (BG), the organismal components from gut contents, that induced more severe systemic inflammation than BiN without Candida. The co-presentation of LPS and BG in mouse serum enhanced inflammatory responses. As such, LPS with Whole Glucan Particle (WGP, a representative BG) induced more severe macrophage responses than LPS alone as determined by supernatant cytokines and gene expression of downstream signals (NFκB, Malt-1 and Syk). Meanwhile, WGP alone did not induced the responses. In parallel, WGP (with or without LPS), but not LPS alone, accelerated macrophage ATP production (extracellular flux analysis) through the upregulation of genes in mitochondria and glycolysis pathway (using RNA sequencing analysis), without the induction of cell activities. These data indicated a WGP pre-conditioning effect on cell energy augmentation. In conclusion, Candida in BiN mice accelerated gut translocation of BG that augmented cell energy status and enhanced pro-inflammatory macrophage responses. Hence, gut fungi and BG were associated with the enhanced systemic inflammation in acute uremia.
Collapse
|
143
|
Jeong H, Lee C, Cheng C, Chou HC, Yang H, Bae H. Targeting of adipose tissue macrophages by bee venom phospholipase A2 attenuates high-fat diet-induced obesity. Int J Obes (Lond) 2021; 45:1656-1667. [PMID: 33947969 PMCID: PMC8310798 DOI: 10.1038/s41366-021-00823-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Revised: 02/24/2021] [Accepted: 04/08/2021] [Indexed: 12/26/2022]
Abstract
Background/objectives Adipose tissue macrophages (ATMs) exist in either the M1 or M2 form. The anti-inflammatory M2 ATMs accumulate in lean individuals, whereas the pro-inflammatory M1 ATMs accumulate in obese individuals. Bee venom phospholipase A2 (bvPLA2), a major component in honeybee (Apis mellifera) venom, exerts potent anti-inflammatory effects via interactions with regulatory T cells (Treg) and macrophages. This study investigated the effects of bvPLA2 on a high-fat diet (HFD)-induced obesity in mice. Subjects/methods For in vivo experiments, male C57BL/6, CD206-deficient, and Treg-depleted mice models were fed either a normal diet 41.86 kJ (ND, 10 kcal% fat) or high-fat diet 251.16 kJ (HFD, 60 kcal% fat). Each group was i.p. injected with PBS or bvPLA2 (0.5 mg/kg) every 3 days for 11 weeks. Body weight and food intake were measured weekly. Histological changes in the white adipose tissue (WAT), liver, and kidney as well as the immune phenotypes of the WAT were examined. Immune cells, cytokines, and lipid profiles were also evaluated. The direct effects of bvPLA2 on 3T3-L1 pre-adipocytes and bone marrow-derived macrophages were measured in vitro. Results bvPLA2 markedly decreased bodyweight in HFD-fed mice. bvPLA2 treatment also decreased lipid accumulation in the liver and reduced kidney inflammation in the mice. It was confirmed that bvPLA2 exerted immunomodulatory effects through the CD206 receptor. In addition, bvPLA2 decreased M1 ATM and alleviated the M1/M2 imbalance in vivo. However, bvPLA2 did not directly inhibit adipogenesis in the 3T3-L1 adipose cells in vitro. Conclusions bvPLA2 is a potential therapeutic strategy for the management of obesity by regulating adipose tissue macrophage homeostasis.
Collapse
Affiliation(s)
- Hyunju Jeong
- Department of Physiology, College of Korean Medicine, Kyung Hee University, Seoul, Republic of Korea
| | - Chanju Lee
- Department of Physiology, College of Korean Medicine, Kyung Hee University, Seoul, Republic of Korea
| | - Chenyu Cheng
- Department of Physiology, College of Korean Medicine, Kyung Hee University, Seoul, Republic of Korea
| | - Hung Chun Chou
- Department of Physiology, College of Korean Medicine, Kyung Hee University, Seoul, Republic of Korea
| | - HyeJin Yang
- Department of Physiology, College of Korean Medicine, Kyung Hee University, Seoul, Republic of Korea.
| | - Hyunsu Bae
- Department of Physiology, College of Korean Medicine, Kyung Hee University, Seoul, Republic of Korea.
| |
Collapse
|
144
|
Yang D, Yang L, Cai J, Hu X, Li H, Zhang X, Zhang X, Chen X, Dong H, Nie H, Li Y. A sweet spot for macrophages: Focusing on polarization. Pharmacol Res 2021; 167:105576. [PMID: 33771700 DOI: 10.1016/j.phrs.2021.105576] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/22/2020] [Revised: 03/19/2021] [Accepted: 03/21/2021] [Indexed: 12/21/2022]
Abstract
Macrophages are a type of functionally plastic cells that can create a pro-/anti-inflammatory microenvironment for organs by producing different kinds of cytokines, chemokines, and growth factors to regulate immunity and inflammatory responses. In addition, they can also be induced to adopt different phenotypes in response to extracellular and intracellular signals, a process defined as M1/M2 polarization. Growing evidence indicates that glycobiology is closely associated with this polarization process. In this research, we review studies of the roles of glycosylation, glucose metabolism, and key lectins in the regulation of macrophages function and polarization to provide a new perspective for immunotherapies for multiple diseases.
Collapse
Affiliation(s)
- Depeng Yang
- School of Life Sciences and Technology, Harbin Institute of Technology, Harbin, Heilongjiang 150001, China
| | - Lijun Yang
- School of Life Sciences and Technology, Harbin Institute of Technology, Harbin, Heilongjiang 150001, China
| | - Jialing Cai
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang, Liaoning 110000, China
| | - Xibo Hu
- School of Life Sciences and Technology, Harbin Institute of Technology, Harbin, Heilongjiang 150001, China
| | - Huaxin Li
- School of Life Sciences and Technology, Harbin Institute of Technology, Harbin, Heilongjiang 150001, China
| | - Xiaoqing Zhang
- School of Life Sciences and Technology, Harbin Institute of Technology, Harbin, Heilongjiang 150001, China
| | - Xiaohan Zhang
- School of Life Sciences and Technology, Harbin Institute of Technology, Harbin, Heilongjiang 150001, China
| | - Xinghe Chen
- School of Life Sciences and Technology, Harbin Institute of Technology, Harbin, Heilongjiang 150001, China
| | - Haiyang Dong
- School of Life Sciences and Technology, Harbin Institute of Technology, Harbin, Heilongjiang 150001, China
| | - Huan Nie
- School of Life Sciences and Technology, Harbin Institute of Technology, Harbin, Heilongjiang 150001, China.
| | - Yu Li
- School of Life Sciences and Technology, Harbin Institute of Technology, Harbin, Heilongjiang 150001, China.
| |
Collapse
|
145
|
Saeedifar AM, Mosayebi G, Ghazavi A, Bushehri RH, Ganji A. Macrophage polarization by phytotherapy in the tumor microenvironment. Phytother Res 2021; 35:3632-3648. [DOI: 10.1002/ptr.7058] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2020] [Revised: 12/28/2020] [Accepted: 02/08/2021] [Indexed: 12/16/2022]
Affiliation(s)
- Amir Mohammad Saeedifar
- Department of Immunology & Microbiology, School of Medicine Arak University of Medical Sciences Arak Iran
| | - Ghasem Mosayebi
- Department of Immunology & Microbiology, School of Medicine Arak University of Medical Sciences Arak Iran
- Molecular and Medicine Research Center Arak University of Medical Sciences Arak Iran
| | - Ali Ghazavi
- Department of Immunology & Microbiology, School of Medicine Arak University of Medical Sciences Arak Iran
- Traditional and Complementary Medicine Research Center (TCMRC) Arak University of Medical Sciences Arak Iran
| | - Rouhollah Hemmati Bushehri
- Department of Immunology & Microbiology, School of Medicine Arak University of Medical Sciences Arak Iran
| | - Ali Ganji
- Department of Immunology & Microbiology, School of Medicine Arak University of Medical Sciences Arak Iran
- Molecular and Medicine Research Center Arak University of Medical Sciences Arak Iran
| |
Collapse
|
146
|
Wei (魏彤) T, Gao (高晶) J, Huang (黄程淋) C, Song (宋蓓) B, Sun (孙孟炜) M, Shen (沈伟利) W. SIRT3 (Sirtuin-3) Prevents Ang II (Angiotensin II)-Induced Macrophage Metabolic Switch Improving Perivascular Adipose Tissue Function. Arterioscler Thromb Vasc Biol 2021; 41:714-730. [PMID: 33327751 DOI: 10.1161/atvbaha.120.315337] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
OBJECTIVE Infiltrated macrophages actively promote perivascular adipose tissue remodeling and represent a dominant population in the perivascular adipose tissue microenvironment of hypertensive mice. However, the role of macrophages in initiating metabolic inflammation remains uncertain. SIRT3 (sirtuin-3), a NAD-dependent deacetylase, is sensitive to metabolic status and mediates adaptation responses. In this study, we investigated the role of SIRT3-mediated metabolic shift in regulating NLRP3 (Nod-like receptor family pyrin domain-containing 3) inflammasome activation. Approach and Results: Here, we report that Ang II (angiotensin II) accelerates perivascular adipose tissue inflammation and fibrosis, accompanied by NLRP3 inflammasome activation and IL (interleukin)-1β secretion in myeloid SIRT3 knockout (SIRT3-/-) mice. This effect is associated with adipose tissue mitochondrial dysfunction. In vitro studies indicate that the deletion of SIRT3 in bone marrow-derived macrophages induces IL-1β production by shifting the metabolic phenotype from oxidative phosphorylation to glycolysis. Mechanistically, SIRT3 deacetylates and activates PDHA1 (pyruvate dehydrogenase E1 alpha) at lysine 83, and the loss of SIRT3 leads to PDH activity decrease and lactate accumulation. Knocking down LDHA (lactate dehydrogenase A) or using carnosine, a buffer against lactic acid, attenuates IL-1β secretion. Furthermore, the blockade of IL-1β from macrophages into brown adipocytes restores thermogenic markers and mitochondrial oxygen consumption. Moreover, NLRP3 knockout (NLRP3-/-) mice exhibited reduced IL-1β production while rescuing the mitochondrial function of brown adipocytes and alleviating perivascular adipose tissue fibrosis. CONCLUSIONS SIRT3 represents a potential therapeutic target to attenuate NLRP3-related inflammation. Pharmacological targeting of glycolytic metabolism may represent an effective therapeutic approach.
Collapse
Affiliation(s)
- Tong Wei (魏彤)
- Department of Cardiovascular Medicine, Department of Hypertension, and Department of General Practice, State Key Laboratory of Medical Genomics, Shanghai Key Laboratory of Hypertension, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, China (T.W., J.G., C.H., B.S., W.S.)
| | - Jing Gao (高晶)
- Department of Cardiovascular Medicine, Department of Hypertension, and Department of General Practice, State Key Laboratory of Medical Genomics, Shanghai Key Laboratory of Hypertension, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, China (T.W., J.G., C.H., B.S., W.S.)
| | - Chenglin Huang (黄程淋)
- Department of Cardiovascular Medicine, Department of Hypertension, and Department of General Practice, State Key Laboratory of Medical Genomics, Shanghai Key Laboratory of Hypertension, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, China (T.W., J.G., C.H., B.S., W.S.)
| | - Bei Song (宋蓓)
- Department of Cardiovascular Medicine, Department of Hypertension, and Department of General Practice, State Key Laboratory of Medical Genomics, Shanghai Key Laboratory of Hypertension, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, China (T.W., J.G., C.H., B.S., W.S.)
| | - Mengwei Sun (孙孟炜)
- Department of Cardiovascular Medicine, Department of Hypertension, and Department of General Practice, State Key Laboratory of Medical Genomics, Shanghai Key Laboratory of Hypertension, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, China (T.W., J.G., C.H., B.S., W.S.)
| | - Weili Shen (沈伟利)
- Key Laboratory of State General Administration of Sport, Shanghai Research Institute of Sports
Science, China (M.S.)
| |
Collapse
|
147
|
Tarazona OA, Pourquié O. Exploring the Influence of Cell Metabolism on Cell Fate through Protein Post-translational Modifications. Dev Cell 2021; 54:282-292. [PMID: 32693060 DOI: 10.1016/j.devcel.2020.06.035] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Revised: 06/26/2020] [Accepted: 06/26/2020] [Indexed: 12/30/2022]
Abstract
The connection between cell fate transitions and metabolic shifts is gaining momentum in the study of cell differentiation in embryonic development, adult stem cells, and cancer pathogenesis. Here, we explore how metabolic transitions influence post-translational modifications (PTMs), which play central roles in the activation of transcriptional programs. PTMs can control the function of transcription factors acting as master regulators of cell fate as well as activation or repression of cell identity genes by regulating chromatin state via histone tail modifications. It now becomes clear that cell metabolism is an integral part of the complex landscape of regulatory mechanisms underlying cell differentiation.
Collapse
Affiliation(s)
- Oscar A Tarazona
- Department of Genetics, Blavatnik Institute of Harvard Medical School and Department of Pathology, Brigham and Women's Hospital, Boston, MA, USA; Harvard Stem Cell Institute, Harvard University, Cambridge, MA, USA.
| | - Olivier Pourquié
- Department of Genetics, Blavatnik Institute of Harvard Medical School and Department of Pathology, Brigham and Women's Hospital, Boston, MA, USA; Harvard Stem Cell Institute, Harvard University, Cambridge, MA, USA.
| |
Collapse
|
148
|
Zarei M, Acharya P, Talahalli RR. Ginger and turmeric lipid-solubles attenuate heated oil-induced hepatic inflammation via the downregulation of NF-kB in rats. Life Sci 2021; 265:118856. [PMID: 33278395 DOI: 10.1016/j.lfs.2020.118856] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Revised: 11/21/2020] [Accepted: 11/25/2020] [Indexed: 12/31/2022]
Abstract
PURPOSE Reusing deep-fried vegetable oils multiple times is a common practice to save costs, and their chronic consumption may cause hepatic dysfunction. In this investigation, we assessed the modulatory effects of ginger and turmeric lipid-solubles that may migrate to oils during heating on the hepatic inflammatory response in rats. METHODS Male Wistar rats were fed with; 1) control {native canola (N-CNO) or native sunflower (N-SFO)} oil, 2) heated (heated canola {(H-CNO) or heated sunflower (H-SFO)} oil, and 3) heated oil with ginger or turmeric {heated canola with ginger (H-CNO + GI) or heated canola oil with turmeric (H-CNO + TU), heated sunflower oil with ginger (H-SFO + GI) or heated sunflower oil with turmeric (H-SFO + TU)} for 120 days. Hepatic inflammatory response comprising eicosanoids, cytokines, and NF-kB were assessed. RESULTS Compared to respective controls, feeding heated oils significantly (p < 0.05); 1) increased eicosanoids (PGE2, LTB4, and LTC4) and cytokines (TNF-α, MCP-1, IL-1β, and IL-6), 2) increased nuclear translocation of NF-kB in the liver, and 3) increased the hepatic expression of 5-LOX, COX-2, BLT-1, and EP-4. However, feeding oils heated with ginger or turmeric positively countered the changes induced by consumption of heated oils. CONCLUSIONS Consumption of repeatedly heated oil may cause hepatic dysfunction by inducing inflammatory stress through NF-kB upregulation. Lipid-solubles from ginger and turmeric that may migrate to oil during heating prevent the hepatic inflammatory response triggered by heated oils in rats.
Collapse
Affiliation(s)
- Mehrdad Zarei
- Dept. of Biochemistry, CSIR-Central Food Technological Research Institute, Mysore 570020, Karnataka, India.
| | - Pooja Acharya
- Dept. of Biochemistry, CSIR-Central Food Technological Research Institute, Mysore 570020, Karnataka, India.
| | | |
Collapse
|
149
|
Xiang Y, Miao H. Lipid Metabolism in Tumor-Associated Macrophages. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1316:87-101. [PMID: 33740245 DOI: 10.1007/978-981-33-6785-2_6] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Macrophages are essential components of the immune system in tumors. It can be recruited and educated to two mainly polarized subpopulations (M1-like and M2-like) of tumor-associated macrophages (TAMs) to display anti-tumor or protumor function during the tumor occurrence and progression. Reprogramming of metabolism, especially lipid metabolism, is a typical characteristic of TAMs polarization, which was confirmed recently as a vital target for tumor therapy. However, the relationship between TAMs and lipid metabolism is still obscure in the past decade. In this review, we will first introduce the historical aspects of TAMs, and then discuss the correlation of main lipids (triglycerides, cholesterol, and phospholipids) to TAMs activation and summarize the mechanisms by which lipid metabolism mediated tumor escape the immunological surveillance as well as currently available drugs targeting these mechanisms. We hope that this chapter will give a better understanding of lipid metabolism in TAMs for those who are interested in this field, and lay a foundation to develop novel strategies for tumor therapy by targeting lipid metabolism.
Collapse
Affiliation(s)
- Yuancai Xiang
- Department of Biochemistry and Molecular Biology, Third Military Medical University (Army Medical University), Chongqing, People's Republic of China
| | - Hongming Miao
- Department of Biochemistry and Molecular Biology, Third Military Medical University (Army Medical University), Chongqing, People's Republic of China.
| |
Collapse
|
150
|
Zhang X, Zink F, Hezel F, Vogt J, Wachter U, Wepler M, Loconte M, Kranz C, Hellmann A, Mizaikoff B, Radermacher P, Hartmann C. Metabolic substrate utilization in stress-induced immune cells. Intensive Care Med Exp 2020; 8:28. [PMID: 33336295 PMCID: PMC7746792 DOI: 10.1186/s40635-020-00316-0] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2020] [Accepted: 06/11/2020] [Indexed: 12/21/2022] Open
Abstract
Immune cell activation leads to the acquisition of new functions, such as proliferation, chemotaxis, and cytokine production. These functional changes require continuous metabolic adaption in order to sustain ATP homeostasis for sufficient host defense. The bioenergetic demands are usually met by the interconnected metabolic pathways glycolysis, TCA cycle, and oxidative phosphorylation. Apart from glucose, other sources, such as fatty acids and glutamine, are able to fuel the TCA cycle.Rising evidence has shown that cellular metabolism has a direct effect on the regulation of immune cell functions. Thus, quiescent immune cells maintain a basal metabolic state, which shifts to an accelerated metabolic level upon immune cell activation in order to promote key effector functions.This review article summarizes distinct metabolic signatures of key immune cell subsets from quiescence to activation and demonstrates a methodical concept of how to assess cellular metabolic pathways. It further discusses why metabolic functions are of rising interest for translational research and how they can be affected by the underlying pathophysiological condition and/or therapeutic interventions.
Collapse
Affiliation(s)
- Xiaomin Zhang
- Institut für Anästhesiologische Pathophysiologie und Verfahrensentwicklung, Universitätsklinikum Ulm, Helmholzstraße 8/1, 89081 Ulm, Germany
| | - Fabian Zink
- Institut für Anästhesiologische Pathophysiologie und Verfahrensentwicklung, Universitätsklinikum Ulm, Helmholzstraße 8/1, 89081 Ulm, Germany
| | - Felix Hezel
- Institut für Anästhesiologische Pathophysiologie und Verfahrensentwicklung, Universitätsklinikum Ulm, Helmholzstraße 8/1, 89081 Ulm, Germany
| | - Josef Vogt
- Institut für Anästhesiologische Pathophysiologie und Verfahrensentwicklung, Universitätsklinikum Ulm, Helmholzstraße 8/1, 89081 Ulm, Germany
| | - Ulrich Wachter
- Institut für Anästhesiologische Pathophysiologie und Verfahrensentwicklung, Universitätsklinikum Ulm, Helmholzstraße 8/1, 89081 Ulm, Germany
| | - Martin Wepler
- Institut für Anästhesiologische Pathophysiologie und Verfahrensentwicklung, Universitätsklinikum Ulm, Helmholzstraße 8/1, 89081 Ulm, Germany
- Klinik für Anästhesiologie, Universitätsklinikum Ulm, Ulm, Germany
| | - Maurizio Loconte
- Anesthesia and Intensive Care, San Martino Policlinico Hospital, IRCCS for Oncolocy and Neuroscience, Genoa, Italy
| | - Christine Kranz
- Institut für Analytische und Bioanalytische Chemie, Universität Ulm, Ulm, Germany
| | - Andreas Hellmann
- Institut für Analytische und Bioanalytische Chemie, Universität Ulm, Ulm, Germany
| | - Boris Mizaikoff
- Institut für Analytische und Bioanalytische Chemie, Universität Ulm, Ulm, Germany
| | - Peter Radermacher
- Institut für Anästhesiologische Pathophysiologie und Verfahrensentwicklung, Universitätsklinikum Ulm, Helmholzstraße 8/1, 89081 Ulm, Germany
| | - Clair Hartmann
- Institut für Anästhesiologische Pathophysiologie und Verfahrensentwicklung, Universitätsklinikum Ulm, Helmholzstraße 8/1, 89081 Ulm, Germany
- Klinik für Anästhesiologie, Universitätsklinikum Ulm, Ulm, Germany
| |
Collapse
|