1
|
Gao G, Cai L, Fan Y, Aroche Ginarte R, Li Y, Sun W, Jiang X, Li X, Pi Y. Effects of Different Hemicellulose Components on Fermentation Kinetics and Microbial Composition in Fecal Inoculum from Suckling Piglets In Vit ro. ACS OMEGA 2025; 10:9120-9131. [PMID: 40092836 PMCID: PMC11904680 DOI: 10.1021/acsomega.4c08848] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Revised: 02/08/2025] [Accepted: 02/19/2025] [Indexed: 03/19/2025]
Abstract
This study investigated the fermentation characteristics of different hemicellulose components using a fecal inoculum derived from suckling piglets. The results showed that after 60 h of fermentation, the arabinogalactan (Ara-gal), glucomannan (Glu-man), galactomannan (Gal-man), and mannan (Man) groups exhibited similar levels of gas production, which were higher than those of the β-glucan (β-Glu) group. The β-Glu group had the lowest pH value. After 48 h of fermentation, the Ara-gal group had the highest microbial crude protein content and the lowest ammonia nitrogen content. The Glu-man, Gal-man, and Man groups produced similar amounts of acetate, propionate, and total short-chain fatty acids (SCFAs), which were higher than those in the Ara-gal and β-Glu groups. Furthermore, the Man and Ara-gal groups showed the highest butyrate production. Significant differences in the microbial community composition were observed among the groups. Correlation analyses further revealed that the abundance of specific bacteria, such as Prevotella_9 and Parabacteroides, was closely related to the production of acetate, propionate, and butyrate. These results suggest that Glu-man, Gal-man, and Man undergo rapid fermentation, with Ara-gal following, while β-Glu ferments the slowest. The distinct fiber compositions and fermentation properties of different hemicellulose components significantly influence the microbial composition and SCFA production. Our findings offer valuable theoretical insights for selecting fiber components in the diets of suckling piglets and potentially in infants.
Collapse
Affiliation(s)
- Ge Gao
- Key Laboratory
of Feed Biotechnology of Ministry of Agriculture and Rural Affairs,
Institute of Feed Research, Chinese Academy
of Agricultural Sciences, Beijing 100081, China
| | - Long Cai
- Key Laboratory
of Feed Biotechnology of Ministry of Agriculture and Rural Affairs,
Institute of Feed Research, Chinese Academy
of Agricultural Sciences, Beijing 100081, China
| | - Yuyang Fan
- Key Laboratory
of Feed Biotechnology of Ministry of Agriculture and Rural Affairs,
Institute of Feed Research, Chinese Academy
of Agricultural Sciences, Beijing 100081, China
| | - Roisbel Aroche Ginarte
- Key Laboratory
of Feed Biotechnology of Ministry of Agriculture and Rural Affairs,
Institute of Feed Research, Chinese Academy
of Agricultural Sciences, Beijing 100081, China
- Department
of Animal Husbandry, Faculty of Agricultural Science, University of Granma, Bayamo 85100, Cuba
| | - Yanpin Li
- Key Laboratory
of Feed Biotechnology of Ministry of Agriculture and Rural Affairs,
Institute of Feed Research, Chinese Academy
of Agricultural Sciences, Beijing 100081, China
| | - Wenjuan Sun
- Key Laboratory
of Feed Biotechnology of Ministry of Agriculture and Rural Affairs,
Institute of Feed Research, Chinese Academy
of Agricultural Sciences, Beijing 100081, China
| | - Xianren Jiang
- Key Laboratory
of Feed Biotechnology of Ministry of Agriculture and Rural Affairs,
Institute of Feed Research, Chinese Academy
of Agricultural Sciences, Beijing 100081, China
| | - Xilong Li
- Key Laboratory
of Feed Biotechnology of Ministry of Agriculture and Rural Affairs,
Institute of Feed Research, Chinese Academy
of Agricultural Sciences, Beijing 100081, China
| | - Yu Pi
- Key Laboratory
of Feed Biotechnology of Ministry of Agriculture and Rural Affairs,
Institute of Feed Research, Chinese Academy
of Agricultural Sciences, Beijing 100081, China
| |
Collapse
|
2
|
Yang Y, Pei T, Liu C, Cao M, Hu X, Yuan J, Chen F, Guo B, Hong Y, Liu J, Li B, Li X, Wang H. Glutamine metabolic competition drives immunosuppressive reprogramming of intratumour GPR109A + myeloid cells to promote liver cancer progression. Gut 2025; 74:255-269. [PMID: 38981667 DOI: 10.1136/gutjnl-2024-332429] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/15/2024] [Accepted: 06/29/2024] [Indexed: 07/11/2024]
Abstract
OBJECTIVE The metabolic characteristics of liver cancer drive considerable hurdles to immune cells function and cancer immunotherapy. However, how metabolic reprograming in the tumour microenvironment impairs the antitumour immune response remains unclear. DESIGN Human samples and multiple murine models were employed to evaluate the correlation between GPR109A and liver cancer progression. GPR109A knockout mice, immune cells depletion and primary cell coculture models were used to determine the regulation of GPR109A on tumour microenvironment and identify the underlying mechanism responsible for the formation of intratumour GPR109A+myeloid cells. RESULTS We demonstrate that glutamine shortage in liver cancer tumour microenvironment drives an immunosuppressive GPR109A+myeloid cells infiltration, leading to the evasion of immune surveillance. Blockade of GPR109A decreases G-MDSCs and M2-like TAMs abundance to trigger the antitumour responses of CD8+ T cells and further improves the immunotherapy efficacy against liver cancer. Mechanistically, tumour cells and tumour-infiltrated myeloid cells compete for glutamine uptake via the transporter SLC1A5 to control antitumour immunity, which disrupts the endoplasmic reticulum (ER) homoeostasis and induces unfolded protein response of myeloid cells to promote GPR109A expression through IRE1α/XBP1 pathway. The restriction of glutamine uptake in liver cancer cells, as well as the blockade of IRE1α/XBP1 signalling or glutamine supplementation, can eliminate the immunosuppressive effects of GPR109A+ myeloid cells and slow down tumour progression. CONCLUSION Our findings identify the immunometabolic crosstalk between liver cancer cells and myeloid cells facilitates tumour progression via a glutamine metabolism/ER stress/GPR109A axis, suggesting that GPR109A can be exploited as an immunometabolic checkpoint and putative target for cancer treatment.
Collapse
Affiliation(s)
- Yang Yang
- State Key Laboratory of Systems Medicine for Cancer, Center for Single-Cell Omics, School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Tianduo Pei
- State Key Laboratory of Systems Medicine for Cancer, Center for Single-Cell Omics, School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Chaobao Liu
- State Key Laboratory of Systems Medicine for Cancer, Center for Single-Cell Omics, School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Mingtao Cao
- State Key Laboratory of Systems Medicine for Cancer, Center for Single-Cell Omics, School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiaolin Hu
- State Key Laboratory of Systems Medicine for Cancer, Center for Single-Cell Omics, School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jie Yuan
- State Key Laboratory of Systems Medicine for Cancer, Center for Single-Cell Omics, School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Fengqian Chen
- State Key Laboratory of Systems Medicine for Cancer, Center for Single-Cell Omics, School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Bao Guo
- State Key Laboratory of Systems Medicine for Cancer, Center for Single-Cell Omics, School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yuemei Hong
- State Key Laboratory of Systems Medicine for Cancer, Center for Single-Cell Omics, School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jibin Liu
- Institute of Oncology, Affiliated Tumor Hospital of Nantong University, Nantong, China
| | - Bin Li
- Biliary Tract Surgery Department I, Eastern Hepatobiliary Surgery Hospital, Secondary Military Medicine University, Shanghai, China
| | - Xiaoguang Li
- State Key Laboratory of Systems Medicine for Cancer, Center for Single-Cell Omics, School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Hui Wang
- State Key Laboratory of Systems Medicine for Cancer, Center for Single-Cell Omics, School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
3
|
Richardson JC, Higgins GA, Upton N, Massey P, Cunningham M, Wilson S, Holenz J, Taylor C, Lavrov A, Lin H, Matsuoka Y, Brown AJ. The hydroxycarboxylic acid receptor HCA2 is required for the protective effect of ketogenic diet in epilepsy. Pharmacol Res Perspect 2024; 12:e70026. [PMID: 39439218 PMCID: PMC11496569 DOI: 10.1002/prp2.70026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2024] [Revised: 09/19/2024] [Accepted: 09/24/2024] [Indexed: 10/25/2024] Open
Abstract
One third of epilepsy patients are resistant to treatment with current anti-seizure medications. The ketogenic diet is used to treat some forms of refractory epilepsy, but the mechanism of its action has not yet been elucidated. In this study, we aimed to investigate whether the hydroxycarboxylic acid receptor 2 (HCA2), a known immunomodulatory receptor, plays a role in mediating the protective effect of this diet. We demonstrate for the first time that selective agonists at this receptor can directly reduce seizures in animal models. Agonists also reduce network activity in rodent and human brain slices. Ketogenic diet is known to increase circulating levels of endogenous HCA2 agonists, and we show that the effect of ketogenic diet in reducing seizures in the 6 Hz seizure model is negated in HCA2-deficient mice. Our data support the potential of HCA2 as a target for the treatment of epilepsy and potentially for neurodegenerative diseases.
Collapse
Affiliation(s)
| | | | | | - Peter Massey
- Institute of NeuroscienceUniversity of NewcastleNewcastleUK
| | - Mark Cunningham
- Institute of NeuroscienceUniversity of NewcastleNewcastleUK
- Discipline of Physiology, School of MedicineTrinity College DublinDublin 2Ireland
| | - Steve Wilson
- In vitro and in vivo TranslationGlaxoSmithKline R&D LtdStevenageUK
| | - Joerg Holenz
- Neurosciences Therapeutic Area UnitGlaxoSmithKline R&D LtdUpper ProvidencePennsylvaniaUSA
| | | | - Arseniy Lavrov
- Neurosciences Therapeutic Area UnitGlaxoSmithKline R&D LtdStockley ParkUK
| | - Hong Lin
- Neurosciences Therapeutic Area UnitGlaxoSmithKline R&D LtdUpper ProvidencePennsylvaniaUSA
| | - Yasuji Matsuoka
- Neurosciences Therapeutic Area UnitGlaxoSmithKline R&D LtdUpper ProvidencePennsylvaniaUSA
| | | |
Collapse
|
4
|
Kim J, Choi C. Orphan GPCRs in Neurodegenerative Disorders: Integrating Structural Biology and Drug Discovery Approaches. Curr Issues Mol Biol 2024; 46:11646-11664. [PMID: 39451571 PMCID: PMC11505999 DOI: 10.3390/cimb46100691] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Revised: 10/11/2024] [Accepted: 10/17/2024] [Indexed: 10/26/2024] Open
Abstract
Neurodegenerative disorders, particularly Alzheimer's and Parkinson's diseases, continue to challenge modern medicine despite therapeutic advances. Orphan G-protein-coupled receptors (GPCRs) have emerged as promising targets in the central nervous system, offering new avenues for drug development. This review focuses on the structural biology of orphan GPCRs implicated in these disorders, providing a comprehensive analysis of their molecular architecture and functional mechanisms. We examine recent breakthroughs in structural determination techniques, such as cryo-electron microscopy and X-ray crystallography, which have elucidated the intricate conformations of these receptors. The review highlights how structural insights inform our understanding of orphan GPCR activation, ligand binding and signaling pathways. By integrating structural data with molecular pharmacology, we explore the potential of structure-guided approaches in developing targeted therapeutics toward orphan GPCRs. This structural-biology-centered perspective aims to deepen our comprehension of orphan GPCRs and guide future drug discovery efforts in neurodegenerative disorders.
Collapse
Affiliation(s)
- Jinuk Kim
- Department of Biological Sciences, Seoul National University, Seoul 08826, Republic of Korea;
| | | |
Collapse
|
5
|
Datta S, Pasham S, Inavolu S, Boini KM, Koka S. Role of Gut Microbial Metabolites in Cardiovascular Diseases-Current Insights and the Road Ahead. Int J Mol Sci 2024; 25:10208. [PMID: 39337693 PMCID: PMC11432476 DOI: 10.3390/ijms251810208] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Revised: 09/18/2024] [Accepted: 09/19/2024] [Indexed: 09/30/2024] Open
Abstract
Cardiovascular diseases (CVDs) are the leading cause of premature morbidity and mortality globally. The identification of novel risk factors contributing to CVD onset and progression has enabled an improved understanding of CVD pathophysiology. In addition to the conventional risk factors like high blood pressure, diabetes, obesity and smoking, the role of gut microbiome and intestinal microbe-derived metabolites in maintaining cardiovascular health has gained recent attention in the field of CVD pathophysiology. The human gastrointestinal tract caters to a highly diverse spectrum of microbes recognized as the gut microbiota, which are central to several physiologically significant cascades such as metabolism, nutrient absorption, and energy balance. The manipulation of the gut microbial subtleties potentially contributes to CVD, inflammation, neurodegeneration, obesity, and diabetic onset. The existing paradigm of studies suggests that the disruption of the gut microbial dynamics contributes towards CVD incidence. However, the exact mechanistic understanding of such a correlation from a signaling perspective remains elusive. This review has focused upon an in-depth characterization of gut microbial metabolites and their role in varied pathophysiological conditions, and highlights the potential molecular and signaling mechanisms governing the gut microbial metabolites in CVDs. In addition, it summarizes the existing courses of therapy in modulating the gut microbiome and its metabolites, limitations and scientific gaps in our current understanding, as well as future directions of studies involving the modulation of the gut microbiome and its metabolites, which can be undertaken to develop CVD-associated treatment options. Clarity in the understanding of the molecular interaction(s) and associations governing the gut microbiome and CVD shall potentially enable the development of novel druggable targets to ameliorate CVD in the years to come.
Collapse
Affiliation(s)
- Sayantap Datta
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, TX 77204, USA
| | - Sindhura Pasham
- Department of Pharmaceutical Sciences, Irma Lerma College of Pharmacy, Texas A&M University, Kingsville, TX 78363, USA
| | - Sriram Inavolu
- Department of Pharmaceutical Sciences, Irma Lerma College of Pharmacy, Texas A&M University, Kingsville, TX 78363, USA
| | - Krishna M Boini
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, TX 77204, USA
| | - Saisudha Koka
- Department of Pharmaceutical Sciences, Irma Lerma College of Pharmacy, Texas A&M University, Kingsville, TX 78363, USA
| |
Collapse
|
6
|
Yang Y, Pei T, Hu X, Lu Y, Huang Y, Wan T, Liu C, Chen F, Guo B, Hong Y, Ba Q, Li X, Wang H. Dietary vitamin B3 supplementation induces the antitumor immunity against liver cancer via biased GPR109A signaling in myeloid cell. Cell Rep Med 2024; 5:101718. [PMID: 39293389 PMCID: PMC11525019 DOI: 10.1016/j.xcrm.2024.101718] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Revised: 06/23/2024] [Accepted: 08/14/2024] [Indexed: 09/20/2024]
Abstract
The impact of dietary nutrients on tumor immunity remains an area of ongoing investigation, particularly regarding the specific role of vitamins and their mechanism. Here, we demonstrate that vitamin B3 (VB3) induces antitumor immunity against liver cancer through biased GPR109A axis in myeloid cell. Nutritional epidemiology studies suggest that higher VB3 intake reduces liver cancer risk. VB3 supplementation demonstrates antitumor efficacy in multiple mouse models through alleviating the immunosuppressive tumor microenvironment (TME) mediated by tumor-infiltrating myeloid cell, thereby augmenting effectiveness of immunotherapy or targeted therapy in a CD8+ T cell-dependent manner. Mechanically, the TME induces aberrant GPR109A/nuclear factor κB (NF-κB) activation in myeloid cell to shape the immunosuppressive TME. In contrast, VB3 activates β-Arrestin-mediated GPR109A degradation and NF-κB inhibition to suppress the immunosuppressive polarization of myeloid cell, thereby activating the cytotoxic function of CD8+ T cell. Overall, these results expand the understanding of how vitamins regulate the TME, suggesting that dietary VB3 supplementation is an adjunctive treatment for liver cancer.
Collapse
Affiliation(s)
- Yang Yang
- State Key Laboratory of Systems Medicine for Cancer, Center for Single-Cell Omics, School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Tianduo Pei
- State Key Laboratory of Systems Medicine for Cancer, Center for Single-Cell Omics, School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Xiaolin Hu
- State Key Laboratory of Systems Medicine for Cancer, Center for Single-Cell Omics, School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Yu Lu
- State Key Laboratory of Systems Medicine for Cancer, Center for Single-Cell Omics, School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Yanqiu Huang
- State Key Laboratory of Systems Medicine for Cancer, Center for Single-Cell Omics, School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Tingya Wan
- State Key Laboratory of Systems Medicine for Cancer, Center for Single-Cell Omics, School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Chaobao Liu
- State Key Laboratory of Systems Medicine for Cancer, Center for Single-Cell Omics, School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Fengqian Chen
- State Key Laboratory of Systems Medicine for Cancer, Center for Single-Cell Omics, School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Bao Guo
- State Key Laboratory of Systems Medicine for Cancer, Center for Single-Cell Omics, School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Yuemei Hong
- State Key Laboratory of Systems Medicine for Cancer, Center for Single-Cell Omics, School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Qian Ba
- State Key Laboratory of Systems Medicine for Cancer, Center for Single-Cell Omics, School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China.
| | - Xiaoguang Li
- State Key Laboratory of Systems Medicine for Cancer, Center for Single-Cell Omics, School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China.
| | - Hui Wang
- State Key Laboratory of Systems Medicine for Cancer, Center for Single-Cell Omics, School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China.
| |
Collapse
|
7
|
Bejaoui S, Chetoui I, Ghribi F, Belhassen D, Abdallah BB, Fayala CB, Boubaker S, Mili S, Soudani N. Exposure to different cobalt chloride levels produces oxidative stress and lipidomic changes and affects the liver structure of Cyprinus carpio juveniles. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2024; 31:51658-51672. [PMID: 39117974 DOI: 10.1007/s11356-024-34578-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Accepted: 07/26/2024] [Indexed: 08/10/2024]
Abstract
The present investigation was undertaken to evaluate the toxic effects of CoCl2-induced hepatotoxicity and fatty acid changes in juvenile Cyprinus carpio. Fish were divided into six experimental groups in duplicate. The first group served as controls. The second group received the lowest exposure dose at 2.5 µg/L. In the third group, fish were exposed to 25 µg/L of CoCl2. The fourth group was exposed to 50 µg/L of CoCl2. The last two groups were exposed to the highest doses, 100 and 500 µg/L of CoCl2. Total antioxidant activities were estimated using a colorimetric method. Liver fatty acid compositions were analyzed by high-performance gas chromatography (GC). Hepatopathy was identified through microscopic analysis. Exposure of C. carpio to CoCl2 resulted in hepatotoxicity, indicated by increased levels of malondialdehyde (MDA), hydrogen peroxide (H2O2), protein carbonyls (PCO), and alterations in the ferric reducing antioxidant power system (FRAP). Superoxide dismutase (SOD), glutathione-S-transferase (GST), glutathione peroxidase (GPx), reduced glutathione (GSH), metallothioneins (MTs), and low thiol levels (L-SH) significantly increased, particularly under exposure to the highest CoCl2 doses (100 and 500 µg/L). Acetylcholinesterase activity decreased significantly in C. carpio exposed to graded CoCl2 doses. Additionally, there was a decrease in polyunsaturated fatty acids (PUFA), primarily n-3 PUFA, docosahexaenoic acid (DHA), and eicosapentaenoic acid (EPA), while an increase in monounsaturated (MUFA) and saturated fatty acids (SFA), including palmitic (C16:0), stearic (C18:0), palmitoleic (C16:1), and oleic (C18:1) acids, was observed. Histopathological examination of the liver confirmed hepatopathy revealing characteristic tissue changes such as leucocyte infiltration, hepatic cell membrane degradation, vacuolization, and lipid inclusions. The study provided ethnophysiology insights into the responses of C. carpio to CoCl2-induced oxidative stress and lipidomic alteration, underscoring its potential as a bioindicator for assessing environmental impacts and metal contamination.
Collapse
Affiliation(s)
- Safa Bejaoui
- Laboratory of Ecology, Biology and Physiology of Aquatic Organisms, Department of Biology, Faculty of Science of Tunis, University of Tunis El Manar, 2092, Tunis, Tunisia.
| | - Imene Chetoui
- Laboratory of Ecology, Biology and Physiology of Aquatic Organisms, Department of Biology, Faculty of Science of Tunis, University of Tunis El Manar, 2092, Tunis, Tunisia
| | - Feriel Ghribi
- Laboratory of Ecology, Biology and Physiology of Aquatic Organisms, Department of Biology, Faculty of Science of Tunis, University of Tunis El Manar, 2092, Tunis, Tunisia
- High Institute of Aquaculture and Fishing of Bizerte, BP15, 7080, Menzel Jemil, Tunisia
| | - Dalya Belhassen
- Laboratory of Ecology, Biology and Physiology of Aquatic Organisms, Department of Biology, Faculty of Science of Tunis, University of Tunis El Manar, 2092, Tunis, Tunisia
| | - Boutheina Ben Abdallah
- Laboratory of Ecology, Biology and Physiology of Aquatic Organisms, Department of Biology, Faculty of Science of Tunis, University of Tunis El Manar, 2092, Tunis, Tunisia
| | - Chaima Ben Fayala
- Unit of Pathological and Experimental Human Anatomy, Institute of Pasteur of Tunis, 1002, Tunis-Belvedere, Tunisia
| | - Samir Boubaker
- Unit of Pathological and Experimental Human Anatomy, Institute of Pasteur of Tunis, 1002, Tunis-Belvedere, Tunisia
| | - Sami Mili
- High Institute of Aquaculture and Fishing of Bizerte, BP15, 7080, Menzel Jemil, Tunisia
| | - Nejla Soudani
- Laboratory of Ecology, Biology and Physiology of Aquatic Organisms, Department of Biology, Faculty of Science of Tunis, University of Tunis El Manar, 2092, Tunis, Tunisia
| |
Collapse
|
8
|
Sui Y, Feng X, Ma Y, Zou Y, Liu Y, Huang J, Zhu X, Wang J. BHBA attenuates endoplasmic reticulum stress-dependent neuroinflammation via the gut-brain axis in a mouse model of heat stress. CNS Neurosci Ther 2024; 30:e14840. [PMID: 38973202 PMCID: PMC11228358 DOI: 10.1111/cns.14840] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2024] [Revised: 06/12/2024] [Accepted: 06/25/2024] [Indexed: 07/09/2024] Open
Abstract
BACKGROUND Heat stress (HS) commonly occurs as a severe pathological response when the body's sensible temperature exceeds its thermoregulatory capacity, leading to the development of chronic brain inflammation, known as neuroinflammation. Emerging evidence suggests that HS leads to the disruption of the gut microbiota, whereas abnormalities in the gut microbiota have been demonstrated to affect neuroinflammation. However, the mechanisms underlying the effects of HS on neuroinflammation are poorly studied. Meanwhile, effective interventions have been unclear. β-Hydroxybutyric acid (BHBA) has been found to have neuroprotective and anti-inflammatory properties in previous studies. This study aims to explore the modulatory effects of BHBA on neuroinflammation induced by HS and elucidate the underlying molecular mechanisms. METHODS An in vivo and in vitro model of HS was constructed under the precondition of BHBA pretreatment. The modulatory effects of BHBA on HS-induced neuroinflammation were explored and the underlying molecular mechanisms were elucidated by flow cytometry, WB, qPCR, immunofluorescence staining, DCFH-DA fluorescent probe assay, and 16S rRNA gene sequencing of colonic contents. RESULTS Heat stress was found to cause gut microbiota disruption in HS mouse models, and TM7 and [Previotella] spp. may be the best potential biomarkers for assessing the occurrence of HS. Fecal microbiota transplantation associated with BHBA effectively reversed the disruption of gut microbiota in HS mice. Moreover, BHBA may inhibit microglia hyperactivation, suppress neuroinflammation (TNF-α, IL-1β, and IL-6), and reduce the expression of cortical endoplasmic reticulum stress (ERS) markers (GRP78 and CHOP) mainly through its modulatory effects on the gut microbiota (TM7, Lactobacillus spp., Ruminalococcus spp., and Prevotella spp.). In vitro experiments revealed that BHBA (1 mM) raised the expression of the ERS marker GRP78, enhanced cellular activity, and increased the generation of reactive oxygen species (ROS) and anti-inflammatory cytokines (IL-10), while also inhibiting HS-induced apoptosis, ROS production, and excessive release of inflammatory cytokines (TNF-α and IL-1β) in mouse BV2 cells. CONCLUSION β-Hydroxybutyric acid may be an effective agent for preventing neuroinflammation in HS mice, possibly due to its ability to inhibit ERS and subsequent microglia neuroinflammation via the gut-brain axis. These findings lay the groundwork for future research and development of BHBA as a preventive drug for HS and provide fresh insights into techniques for treating neurological illnesses by modifying the gut microbiota.
Collapse
Affiliation(s)
- Yuzhen Sui
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China
| | - Xiao Feng
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China
| | - Yue Ma
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China
| | - Yimeng Zou
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China
| | - Yanli Liu
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China
| | - Jian Huang
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China
| | - Xiaoyan Zhu
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China
| | - Jianguo Wang
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China
| |
Collapse
|
9
|
Tsuruta H, Yamahara K, Yasuda-Yamahara M, Kume S. Emerging Pathophysiological Roles of Ketone Bodies. Physiology (Bethesda) 2024; 39:0. [PMID: 38260943 DOI: 10.1152/physiol.00031.2023] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Revised: 01/15/2024] [Accepted: 01/18/2024] [Indexed: 01/24/2024] Open
Abstract
The discovery of insulin approximately a century ago greatly improved the management of diabetes, including many of its life-threatening acute complications like ketoacidosis. This breakthrough saved many lives and extended the healthy lifespan of many patients with diabetes. However, there is still a negative perception of ketone bodies stemming from ketoacidosis. Originally, ketone bodies were thought of as a vital source of energy during fasting and exercise. Furthermore, in recent years, research on calorie restriction and its potential impact on extending healthy lifespans, as well as studies on ketone bodies, have gradually led to a reevaluation of the significance of ketone bodies in promoting longevity. Thus, in this review, we discuss the emerging and hidden roles of ketone bodies in various organs, including the heart, kidneys, skeletal muscles, and brain, as well as their potential impact on malignancies and lifespan.
Collapse
Affiliation(s)
- Hiroaki Tsuruta
- Department of Medicine, Shiga University of Medical Science, Seta, Otsu, Shiga, Japan
| | - Kosuke Yamahara
- Department of Medicine, Shiga University of Medical Science, Seta, Otsu, Shiga, Japan
| | - Mako Yasuda-Yamahara
- Department of Medicine, Shiga University of Medical Science, Seta, Otsu, Shiga, Japan
| | - Shinji Kume
- Department of Medicine, Shiga University of Medical Science, Seta, Otsu, Shiga, Japan
| |
Collapse
|
10
|
Liu J, Chen Q, Su R. Interplay of human gastrointestinal microbiota metabolites: Short-chain fatty acids and their correlation with Parkinson's disease. Medicine (Baltimore) 2024; 103:e37960. [PMID: 38669388 PMCID: PMC11049718 DOI: 10.1097/md.0000000000037960] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/28/2024] [Accepted: 03/29/2024] [Indexed: 04/28/2024] Open
Abstract
Short-chain fatty acids (SCFAs) are, the metabolic byproducts of intestinal microbiota that, are generated through anaerobic fermentation of undigested dietary fibers. SCFAs play a pivotal role in numerous physiological functions within the human body, including maintaining intestinal mucosal health, modulating immune functions, and regulating energy metabolism. In recent years, extensive research evidence has indicated that SCFAs are significantly involved in the onset and progression of Parkinson disease (PD). However, the precise mechanisms remain elusive. This review comprehensively summarizes the progress in understanding how SCFAs impact PD pathogenesis and the underlying mechanisms. Primarily, we delve into the synthesis, metabolism, and signal transduction of SCFAs within the human body. Subsequently, an analysis of SCFA levels in patients with PD is presented. Furthermore, we expound upon the mechanisms through which SCFAs induce inflammatory responses, oxidative stress, abnormal aggregation of alpha-synuclein, and the intricacies of the gut-brain axis. Finally, we provide a critical analysis and explore the potential therapeutic role of SCFAs as promising targets for treating PD.
Collapse
Affiliation(s)
- Jiaji Liu
- Inner Mongolia Medical University, Department of Laboratory Medicine, Affiliated Hospital of Inner Mongolia Medical University, Hohhot, China
| | - Qi Chen
- The Third Clinical Medical College of Ningxia Medical University, Ningxia, China
| | - Ruijun Su
- Inner Mongolia Medical University, Department of Laboratory Medicine, Affiliated Hospital of Inner Mongolia Medical University, Hohhot, China
| |
Collapse
|
11
|
Mamedova LK, Krogstad KC, McDonald PO, Pokhrel L, Hua DH, Titgemeyer EC, Bradford BJ. Investigation of HCAR2 antagonists as a potential strategy to modulate bovine leukocytes. J Anim Sci Biotechnol 2024; 15:38. [PMID: 38444010 PMCID: PMC10916251 DOI: 10.1186/s40104-024-00999-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Accepted: 01/17/2024] [Indexed: 03/07/2024] Open
Abstract
BACKGROUND Dairy cows experiencing ketosis after calving suffer greater disease incidence and are at greater risk of leaving the herd. In vitro administration of beta-hydroxybutyric acid (BHBA; the primary blood ketone) has inhibitory effects on the function of bovine leukocytes. BHBA is a ligand of HCAR2 and the activation of these receptors promotes an anti-inflammatory response which may be related with immunosuppression observed in transition dairy cattle. The objective of this study was to identify and test antagonists for HCAR2 in bovine immune cells cultured with BHBA. RESULTS We observed expression of HCAR2 at the protein level within lymphocytes, monocytes, and granulocytes. The proportion of cells expressing HCAR2 tended to be greater in mid-lactation compared to early lactation cows; the increase was a result of increased proportion of T and B cells expressing HCAR2. Stimulation of HCAR2 with niacin or BHBA promoted Ca2+ mobilization in neutrophils and mononuclear cells. Mononuclear cells treated with BHBA had diminished intracellular Ca2+ responses when HCAR2 was knocked down by siRNA silencing, indicating Ca2+ mobilization was mediated by HCAR2 signaling. Two candidate antagonists for HCAR2, synthesized from niacin (NA-1 and NA-5), were tested; monocytes and neutrophils pre-treated with NA-1 and NA-5 had reduced Ca2+ mobilization after incubation with BHBA. Furthermore, NA-5 but not NA-1 prevented BHBA-associated reductions in cyclic AMP. CONCLUSIONS We demonstrated that HCAR2 is present on bovine leukocytes and has greater expression later in lactation. We confirmed that BHBA and niacin derived HCAR2 antagonists alter bovine leukocyte activity. Our results demonstrate that both BHBA and niacin affect bovine leukocyte Ca2+ mobilization in a HCAR2-dependent manner.
Collapse
Affiliation(s)
- Laman K Mamedova
- Department of Animal Science, Michigan State University, East Lansing, Michigan, 48824, USA
- Department of Animal Sciences and Industry, Kansas State University, Manhattan, KS, 66506, USA
| | - Kirby C Krogstad
- Department of Animal Science, Michigan State University, East Lansing, Michigan, 48824, USA
| | - Paiton O McDonald
- Comparative Medicine and Integrative Biology, East Lansing, MI, 48824, USA
| | - Laxman Pokhrel
- Department of Chemistry, Kansas State University, Manhattan, KS, 66506, USA
| | - Duy H Hua
- Department of Chemistry, Kansas State University, Manhattan, KS, 66506, USA
| | - Evan C Titgemeyer
- Department of Animal Sciences and Industry, Kansas State University, Manhattan, KS, 66506, USA
| | - Barry J Bradford
- Department of Animal Science, Michigan State University, East Lansing, Michigan, 48824, USA.
- Department of Animal Sciences and Industry, Kansas State University, Manhattan, KS, 66506, USA.
| |
Collapse
|
12
|
Kumar V, Stewart JH. Obesity, bone marrow adiposity, and leukemia: Time to act. Obes Rev 2024; 25:e13674. [PMID: 38092420 DOI: 10.1111/obr.13674] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Revised: 10/07/2023] [Accepted: 11/13/2023] [Indexed: 02/28/2024]
Abstract
Obesity has taken the face of a pandemic with less direct concern among the general population and scientific community. However, obesity is considered a low-grade systemic inflammation that impacts multiple organs. Chronic inflammation is also associated with different solid and blood cancers. In addition, emerging evidence demonstrates that individuals with obesity are at higher risk of developing blood cancers and have poorer clinical outcomes than individuals in a normal weight range. The bone marrow is critical for hematopoiesis, lymphopoiesis, and myelopoiesis. Therefore, it is vital to understand the mechanisms by which obesity-associated changes in BM adiposity impact leukemia development. BM adipocytes are critical to maintain homeostasis via different means, including immune regulation. However, obesity increases BM adiposity and creates a pro-inflammatory environment to upregulate clonal hematopoiesis and a leukemia-supportive environment. Obesity further alters lymphopoiesis and myelopoiesis via different mechanisms, which dysregulate myeloid and lymphoid immune cell functions mentioned in the text under different sequentially discussed sections. The altered immune cell function during obesity alters hematological malignancies and leukemia susceptibility. Therefore, obesity-induced altered BM adiposity, immune cell generation, and function impact an individual's predisposition and severity of leukemia, which should be considered a critical factor in leukemia patients.
Collapse
Affiliation(s)
- Vijay Kumar
- Department of Surgery, Laboratory of Tumor Immunology and Immunotherapy, Morehouse School of Medicine, Atlanta, Georgia, USA
| | - John H Stewart
- Department of Surgery, Laboratory of Tumor Immunology and Immunotherapy, Morehouse School of Medicine, Atlanta, Georgia, USA
| |
Collapse
|
13
|
Rastegari B, Ghamar Talepoor A, Khosropanah S, Doroudchi M. In Vitro Targeted Delivery of Simvastatin and Niacin to Macrophages Using Mannan-Grafted Magnetite Nanoparticles. ACS OMEGA 2024; 9:658-674. [PMID: 38222576 PMCID: PMC10785661 DOI: 10.1021/acsomega.3c06389] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/27/2023] [Revised: 11/25/2023] [Accepted: 11/29/2023] [Indexed: 01/16/2024]
Abstract
Atherosclerosis, a leading cause of mortality worldwide, involves various subsets of macrophages that contribute to its initiation and progression. Current treatment approaches focus on systemic, long-term administration of cholesterol-lowering antioxidants such as statins and certain vitamins, which unfortunately come with prolonged side effects. To overcome these drawbacks, a mannose-containing magnetic nanoparticle (NP) is introduced as a drug delivery system to specifically target macrophages in vitro using simvastatin or niacin and a combinational therapy approach that reduces local inflammation while avoiding unwanted side effects. The synthesized NPs exhibited superparamagnetic behavior, neutrally charged thin coating with a hydrodynamic size of 77.23 ± 13.90 nm, and a metallic core ranging from 15 to 25 nm. Efficient loading of niacin (87.21%) and simvastatin (75.36%) on the NPs was achieved at respective weights of 20.13 and 5.03 (w/w). In the presence of a mannan hydrolyzing enzyme, 79.51% of simvastatin and 67.23% of niacin were released from the NPs within 90 min, with a leakage rate below 19.22%. Additionally, the coated NPs showed no destructive effect on J774A macrophages up to a concentration of 200 μg/mL. Simvastatin-loaded NPs exhibited a minimal increase in IL-6 expression. The low dosage of simvastatin decreased both IL-6 and ARG1 expressions, while niacin and combined simvastatin/niacin increased the level of ARG1 expression significantly. Toxicity evaluations on human umbilical vein endothelial cells and murine liver cells revealed that free simvastatin administration caused significant toxicity, whereas the encapsulated forms of simvastatin, niacin, and a combination of simvastatin/niacin at equivalent concentrations exhibited no significant toxicity. Hence, the controlled release of the encapsulated form of simvastatin and niacin resulted in the effective modulation of macrophage polarization. The delivery system showed suitability for targeting macrophages to atherosclerotic plaque.
Collapse
Affiliation(s)
- Banafsheh Rastegari
- Diagnostic
Laboratory Sciences and Technology Research Center, School of Paramedical
Sciences, Shiraz University of Medical Sciences, Shiraz 71439-14693, Iran
- Department
of Immunology, School of Medicine, Shiraz
University of Medical Sciences, Shiraz 71348-45794, Iran
| | - Atefe Ghamar Talepoor
- Department
of Immunology, School of Medicine, Shiraz
University of Medical Sciences, Shiraz 71348-45794, Iran
- Immunology
Center for Excellence, School of Medicine, Shiraz University of Medical Sciences, Shiraz 71348-45794, Iran
| | - Shahdad Khosropanah
- Department
of Cardiology, School of Medicine, Shiraz
University of Medical Sciences, Shiraz 71348-45794, Iran
| | - Mehrnoosh Doroudchi
- Department
of Immunology, School of Medicine, Shiraz
University of Medical Sciences, Shiraz 71348-45794, Iran
- Immunology
Center for Excellence, School of Medicine, Shiraz University of Medical Sciences, Shiraz 71348-45794, Iran
| |
Collapse
|
14
|
Macvanin MT, Gluvic ZM, Klisic AN, Manojlovic MS, Suri JS, Rizzo M, Isenovic ER. The Link between miRNAs and PCKS9 in Atherosclerosis. Curr Med Chem 2024; 31:6926-6956. [PMID: 37990898 DOI: 10.2174/0109298673262124231102042914] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Revised: 06/30/2023] [Accepted: 09/11/2023] [Indexed: 11/23/2023]
Abstract
Cardiovascular disease (CDV) represents the major cause of death globally. Atherosclerosis, as the primary cause of CVD, is a chronic immune-inflammatory disorder with complex multifactorial pathophysiology encompassing oxidative stress, enhanced immune-inflammatory cascade, endothelial dysfunction, and thrombosis. An initiating event in atherosclerosis is the subendothelial accumulation of low-density lipoprotein (LDL), followed by the localization of macrophages to fatty deposits on blood vessel walls, forming lipid-laden macrophages (foam cells) that secrete compounds involved in plaque formation. Given the fact that foam cells are one of the key culprits that underlie the pathophysiology of atherosclerosis, special attention has been paid to the investigation of the efficient therapeutic approach to overcome the dysregulation of metabolism of cholesterol in macrophages, decrease the foam cell formation and/or to force its degradation. Proprotein convertase subtilisin/kexin type 9 (PCSK9) is a secretory serine proteinase that has emerged as a significant regulator of the lipid metabolism pathway. PCSK9 activation leads to the degradation of LDL receptors (LDLRs), increasing LDL cholesterol (LDL-C) levels in the circulation. PCSK9 pathway dysregulation has been identified as one of the mechanisms involved in atherosclerosis. In addition, microRNAs (miRNAs) are investigated as important epigenetic factors in the pathophysiology of atherosclerosis and dysregulation of lipid metabolism. This review article summarizes the recent findings connecting the role of PCSK9 in atherosclerosis and the involvement of various miRNAs in regulating the expression of PCSK9-related genes. We also discuss PCSK9 pathway-targeting therapeutic interventions based on PCSK9 inhibition, and miRNA levels manipulation by therapeutic agents.
Collapse
Affiliation(s)
- Mirjana T Macvanin
- Department of Radiobiology and Molecular Genetics, VINČA Institute of Nuclear Sciences, National Institute of the Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - Zoran M Gluvic
- Department of Endocrinology and Diabetes, School of Medicine, University Clinical-Hospital Centre Zemun-Belgrade, Clinic of Internal Medicine, University of Belgrade, Belgrade, Serbia
| | - Aleksandra N Klisic
- Faculty of Medicine, Center for Laboratory Diagnostic, Primary Health Care Center, University of Montenegro, Podgorica, Montenegro
| | - Mia S Manojlovic
- Faculty of Medicine Novi Sad, University of Novi Sad, Novi Sad, Serbia
- Clinic for Endocrinology, Diabetes and Metabolic Disorders, Clinical Center of Vojvodina, Novi Sad, Serbia
| | - Jasjit S Suri
- Stroke Monitoring and Diagnostic Division, Athero- Point™, Roseville, CA95661, USA
| | - Manfredi Rizzo
- Department of Health Promotion, School of Medicine, Mother and Child Care and Medical Specialties (Promise), University of Palermo, Palermo, Italy
| | - Esma R Isenovic
- Department of Radiobiology and Molecular Genetics, VINČA Institute of Nuclear Sciences, National Institute of the Republic of Serbia, University of Belgrade, Belgrade, Serbia
| |
Collapse
|
15
|
Espinoza-Derout J, Arambulo JML, Ramirez-Trillo W, Rivera JC, Hasan KM, Lao CJ, Jordan MC, Shao XM, Roos KP, Sinha-Hikim AP, Friedman TC. The lipolysis inhibitor acipimox reverses the cardiac phenotype induced by electronic cigarettes. Sci Rep 2023; 13:18239. [PMID: 37880325 PMCID: PMC10600141 DOI: 10.1038/s41598-023-44082-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Accepted: 10/03/2023] [Indexed: 10/27/2023] Open
Abstract
Electronic cigarettes (e-cigarettes) are a prevalent alternative to conventional nicotine cigarettes among smokers and people who have never smoked. Increased concentrations of serum free fatty acids (FFAs) are crucial in generating lipotoxicity. We studied the effects of acipimox, an antilipolytic drug, on e-cigarette-induced cardiac dysfunction. C57BL/6J wild-type mice on high fat diet were treated with saline, e-cigarette with 2.4% nicotine [e-cigarette (2.4%)], and e-cigarette (2.4%) plus acipimox for 12 weeks. Fractional shortening and ejection fraction were diminished in mice exposed to e-cigarettes (2.4%) compared with saline and acipimox-treated mice. Mice exposed to e-cigarette (2.4%) had increased circulating levels of inflammatory cytokines and FFAs, which were diminished by acipimox. Gene Set Enrichment Analysis revealed that e-cigarette (2.4%)-treated mice had gene expression changes in the G2/M DNA damage checkpoint pathway that was normalized by acipimox. Accordingly, we showed that acipimox suppressed the nuclear localization of phospho-p53 induced by e-cigarette (2.4%). Additionally, e-cigarette (2.4%) increased the apurinic/apyrimidinic sites, a marker of oxidative DNA damage which was normalized by acipimox. Mice exposed to e-cigarette (2.4%) had increased cardiac Heme oxygenase 1 protein levels and 4-hydroxynonenal (4-HNE). These markers of oxidative stress were decreased by acipimox. Therefore, inhibiting lipolysis with acipimox normalizes the physiological changes induced by e-cigarettes and the associated increase in inflammatory cytokines, oxidative stress, and DNA damage.
Collapse
Affiliation(s)
- Jorge Espinoza-Derout
- Division of Endocrinology, Metabolism and Molecular Medicine, Department of Internal Medicine, Charles R. Drew University of Medicine and Science, 1731 E. 120th Street, Los Angeles, CA, 90059, USA.
- Departments of Physiology, Medicine, and Neurobiology, David Geffen School of Medicine at University of California, Los Angeles, Los Angeles, CA, 90095, USA.
| | - Jose Mari Luis Arambulo
- Division of Endocrinology, Metabolism and Molecular Medicine, Department of Internal Medicine, Charles R. Drew University of Medicine and Science, 1731 E. 120th Street, Los Angeles, CA, 90059, USA
| | - William Ramirez-Trillo
- Division of Endocrinology, Metabolism and Molecular Medicine, Department of Internal Medicine, Charles R. Drew University of Medicine and Science, 1731 E. 120th Street, Los Angeles, CA, 90059, USA
| | - Juan Carlos Rivera
- Division of Endocrinology, Metabolism and Molecular Medicine, Department of Internal Medicine, Charles R. Drew University of Medicine and Science, 1731 E. 120th Street, Los Angeles, CA, 90059, USA
| | - Kamrul M Hasan
- Division of Endocrinology, Metabolism and Molecular Medicine, Department of Internal Medicine, Charles R. Drew University of Medicine and Science, 1731 E. 120th Street, Los Angeles, CA, 90059, USA
- Departments of Physiology, Medicine, and Neurobiology, David Geffen School of Medicine at University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - Candice J Lao
- Division of Endocrinology, Metabolism and Molecular Medicine, Department of Internal Medicine, Charles R. Drew University of Medicine and Science, 1731 E. 120th Street, Los Angeles, CA, 90059, USA
- Departments of Physiology, Medicine, and Neurobiology, David Geffen School of Medicine at University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - Maria C Jordan
- Departments of Physiology, Medicine, and Neurobiology, David Geffen School of Medicine at University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - Xuesi M Shao
- Division of Endocrinology, Metabolism and Molecular Medicine, Department of Internal Medicine, Charles R. Drew University of Medicine and Science, 1731 E. 120th Street, Los Angeles, CA, 90059, USA
- Departments of Physiology, Medicine, and Neurobiology, David Geffen School of Medicine at University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - Kenneth P Roos
- Departments of Physiology, Medicine, and Neurobiology, David Geffen School of Medicine at University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - Amiya P Sinha-Hikim
- Division of Endocrinology, Metabolism and Molecular Medicine, Department of Internal Medicine, Charles R. Drew University of Medicine and Science, 1731 E. 120th Street, Los Angeles, CA, 90059, USA
- Departments of Physiology, Medicine, and Neurobiology, David Geffen School of Medicine at University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - Theodore C Friedman
- Division of Endocrinology, Metabolism and Molecular Medicine, Department of Internal Medicine, Charles R. Drew University of Medicine and Science, 1731 E. 120th Street, Los Angeles, CA, 90059, USA
- Departments of Physiology, Medicine, and Neurobiology, David Geffen School of Medicine at University of California, Los Angeles, Los Angeles, CA, 90095, USA
| |
Collapse
|
16
|
Masse KE, Lu VB. Short-chain fatty acids, secondary bile acids and indoles: gut microbial metabolites with effects on enteroendocrine cell function and their potential as therapies for metabolic disease. Front Endocrinol (Lausanne) 2023; 14:1169624. [PMID: 37560311 PMCID: PMC10407565 DOI: 10.3389/fendo.2023.1169624] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/19/2023] [Accepted: 07/05/2023] [Indexed: 08/11/2023] Open
Abstract
The gastrointestinal tract hosts the largest ecosystem of microorganisms in the body. The metabolism of ingested nutrients by gut bacteria produces novel chemical mediators that can influence chemosensory cells lining the gastrointestinal tract. Specifically, hormone-releasing enteroendocrine cells which express a host of receptors activated by these bacterial metabolites. This review will focus on the activation mechanisms of glucagon-like peptide-1 releasing enteroendocrine cells by the three main bacterial metabolites produced in the gut: short-chain fatty acids, secondary bile acids and indoles. Given the importance of enteroendocrine cells in regulating glucose homeostasis and food intake, we will also discuss therapies based on these bacterial metabolites used in the treatment of metabolic diseases such as diabetes and obesity. Elucidating the mechanisms gut bacteria can influence cellular function in the host will advance our understanding of this fundamental symbiotic relationship and unlock the potential of harnessing these pathways to improve human health.
Collapse
Affiliation(s)
| | - Van B. Lu
- Department of Physiology and Pharmacology, University of Western Ontario, London, ON, Canada
| |
Collapse
|
17
|
Perrone M, Pagano M, Belardo C, Ricciardi F, Ricciardi F, Fusco A, Trotta MC, Infantino R, Gargano F, Parente A, Giacca R, Pieretti G, Luongo L, Maione S, Boccella S, Guida F. Potential role of the hydroxyl carboxylic acid receptor type 2 (HCAR2) in microglia pathophysiology: A possible cross-talk with C-X-C chemokine receptor 1 (CXCR1). Neuropharmacology 2023; 228:109456. [PMID: 36796675 DOI: 10.1016/j.neuropharm.2023.109456] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Revised: 01/27/2023] [Accepted: 02/05/2023] [Indexed: 02/16/2023]
Abstract
Following insults or injury, microglia cells are activated contributing to the cytotoxic response or by promoting an immune-mediated damage resolution. Microglia cells express HCA2R, a hydroxy carboxylic acid (HCA) receptor, which has been shown to mediate neuroprotective and anti-inflammatory effects. In this study we found that HCAR2 expression levels were increased in cultured rat microglia cells after Lipopolysaccharide (LPS) exposure. In a similar fashion, the treatment with MK 1903, a potent full agonist of HCAR2, increased the receptor protein levels. Moreover, HCAR2 stimulation prevented i) cells viability ii) morphological activation iii) pro/anti-inflammatory mediators production in LPS-treated cells. Likewise, HCAR2 stimulation reduced the proinflammatory mediators mRNA expression induced by neuronal chemokine fractalkine (FKN), a neuronal derived chemokine activating its unique receptor, chemokine receptor 1 (CX3CR1) on microglia surface. Interestingly, electrophysiological recordings in vivo revealed that MK1903 was able to prevent the increase of the nociceptive neurons (NS) firing activity mediated by the spinal FKN application in healthy rats. Collectively, our data demonstrate that HCAR2 is functionally expressed in microglia, by showing its capability to shift microglia toward an anti-inflammatory phenotype. Moreover, we indicated the contribute of HCAR2 in the FKN signaling and suggested a possible HCAR2/CX3CR1 functional interaction. This study paves the way for further investigations aimed at understanding the role HCAR2 as potential target in neuroinflammation-based CNS disorders. This article is part of the Special Issue on "The receptor-receptor interaction as a new target for therapy".
Collapse
Affiliation(s)
- Michela Perrone
- Department of Experimental Medicine, Division of Pharmacology, University of Campania "L. Vanvitelli", Naples, Italy
| | - Martina Pagano
- Department of Experimental Medicine, Division of Pharmacology, University of Campania "L. Vanvitelli", Naples, Italy
| | - Carmela Belardo
- Department of Experimental Medicine, Division of Pharmacology, University of Campania "L. Vanvitelli", Naples, Italy
| | - Flavia Ricciardi
- Department of Experimental Medicine, Division of Pharmacology, University of Campania "L. Vanvitelli", Naples, Italy
| | - Federica Ricciardi
- Department of Experimental Medicine, Division of Pharmacology, University of Campania "L. Vanvitelli", Naples, Italy
| | - Antimo Fusco
- Department of Experimental Medicine, Division of Pharmacology, University of Campania "L. Vanvitelli", Naples, Italy
| | - Maria Consiglia Trotta
- Department of Experimental Medicine, Division of Pharmacology, University of Campania "L. Vanvitelli", Naples, Italy
| | - Rosmara Infantino
- Department of Experimental Medicine, Division of Pharmacology, University of Campania "L. Vanvitelli", Naples, Italy
| | - Francesca Gargano
- Unit of Anaesthesia, Intensive Care and Pain Management, Department of Medicine, Campus Bio-Medico University of Rome, 00128, Rome, Italy
| | - Andrea Parente
- Department of Precision Medicine, Division of Biochemistry, Biophysics and General Pathology, University of Campania "L. Vanvitelli", Naples, Italy
| | - Rosa Giacca
- Department of Precision Medicine, Division of Biochemistry, Biophysics and General Pathology, University of Campania "L. Vanvitelli", Naples, Italy
| | - Gorizio Pieretti
- Multidisciplinary Department of Surgical and Dental Specialities, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Livio Luongo
- Department of Experimental Medicine, Division of Pharmacology, University of Campania "L. Vanvitelli", Naples, Italy.
| | - Sabatino Maione
- Department of Experimental Medicine, Division of Pharmacology, University of Campania "L. Vanvitelli", Naples, Italy.
| | - Serena Boccella
- Department of Experimental Medicine, Division of Pharmacology, University of Campania "L. Vanvitelli", Naples, Italy
| | - Francesca Guida
- Department of Experimental Medicine, Division of Pharmacology, University of Campania "L. Vanvitelli", Naples, Italy
| |
Collapse
|
18
|
Guo W, Gong Q, Zong X, Wu D, Li Y, Xiao H, Song J, Zhang S, Fu S, Feng Z, Zhuang L. GPR109A controls neutrophil extracellular traps formation and improve early sepsis by regulating ROS/PAD4/Cit-H3 signal axis. Exp Hematol Oncol 2023; 12:15. [PMID: 36721229 PMCID: PMC9887879 DOI: 10.1186/s40164-023-00376-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2022] [Accepted: 01/21/2023] [Indexed: 02/01/2023] Open
Abstract
BACKGROUND Neutrophil extracellular traps (NETs) is the key means for neutrophils to resist bacterial invasion. Sepsis is a systemic inflammatory response syndrome caused by infection. METHODS In our study, qRT-PCR was used to detect the gene expression in neutrophils, Western blot was used to detect the protein expression in mouse tissues and neutrophils, flow cytometry was used to detect the purity of neutrophils in the whole blood and immunofluorescence was used to detect the NETs formation. RESULTS In this study, we analyzed the NETs formation in the blood of patients with sepsis. The results showed that a large number of NETs appeared. And the expression of GPR109A in neutrophils of patients with sepsis was significantly up regulated. Then we collected neutrophils from WT mice and GPR109A-/- mice and found that GPR109A knockout could significantly inhibit the early NETs formation of neutrophils. The results also showed that knockout of GPR109A or inhibition of the NETs formation could increase the inflammatory response of liver, spleen, lung and kidney in mice, thus affecting the disease process of sepsis. Then we observed the death of mice in 16 days. The results showed that inhibiting the NETs formation could significantly affect the early mortality of mice, while knocking out GPR109A could directly affect the mortality of the whole period. CONCLUSIONS This study confirmed the regulatory effect of GPR109A on early NETs formation for the first time, and provided a new target for the treatment of sepsis.
Collapse
Affiliation(s)
- Wenjin Guo
- grid.64924.3d0000 0004 1760 5735College of Veterinary Medicine, Jilin University, Changchun, 130062 China
| | - Qian Gong
- grid.13402.340000 0004 1759 700XCollege of Animal Sciences, Zhejiang University, Hangzhou, 310030 China
| | - Xiaofeng Zong
- grid.64924.3d0000 0004 1760 5735College of Veterinary Medicine, Jilin University, Changchun, 130062 China
| | - Dianjun Wu
- grid.64924.3d0000 0004 1760 5735College of Veterinary Medicine, Jilin University, Changchun, 130062 China
| | - Yuhang Li
- grid.64924.3d0000 0004 1760 5735College of Veterinary Medicine, Jilin University, Changchun, 130062 China
| | - Huijie Xiao
- grid.415954.80000 0004 1771 3349Department of Gastrointestinal and Colorectal Surgery, China-Japan Union, Hospital of Jilin University, Changchun, 130033 China
| | - Jie Song
- grid.414252.40000 0004 1761 8894Senior Department of Pediatrics, The Seventh Medical Center of PLA General Hospital, Beijing, China ,grid.414252.40000 0004 1761 8894Institute of Pediatrics, The Seventh Medical Center of PLA General Hospital, Beijing, China ,National Engineering Laboratory for Birth Defects Prevention and Control of Key Technology, Beijing, China ,Beijing Key Laboratory of Pediatric Organ Failure, Beijing, China
| | - Sheng Zhang
- grid.414252.40000 0004 1761 8894Senior Department of Pediatrics, The Seventh Medical Center of PLA General Hospital, Beijing, China ,grid.414252.40000 0004 1761 8894Institute of Pediatrics, The Seventh Medical Center of PLA General Hospital, Beijing, China ,National Engineering Laboratory for Birth Defects Prevention and Control of Key Technology, Beijing, China ,Beijing Key Laboratory of Pediatric Organ Failure, Beijing, China
| | - Shoupeng Fu
- grid.64924.3d0000 0004 1760 5735College of Veterinary Medicine, Jilin University, Changchun, 130062 China
| | - Zhichun Feng
- grid.414252.40000 0004 1761 8894Senior Department of Pediatrics, The Seventh Medical Center of PLA General Hospital, Beijing, China ,grid.414252.40000 0004 1761 8894Institute of Pediatrics, The Seventh Medical Center of PLA General Hospital, Beijing, China ,National Engineering Laboratory for Birth Defects Prevention and Control of Key Technology, Beijing, China ,Beijing Key Laboratory of Pediatric Organ Failure, Beijing, China
| | - Lu Zhuang
- grid.414252.40000 0004 1761 8894Senior Department of Pediatrics, The Seventh Medical Center of PLA General Hospital, Beijing, China ,grid.414252.40000 0004 1761 8894Institute of Pediatrics, The Seventh Medical Center of PLA General Hospital, Beijing, China ,National Engineering Laboratory for Birth Defects Prevention and Control of Key Technology, Beijing, China ,Beijing Key Laboratory of Pediatric Organ Failure, Beijing, China
| |
Collapse
|
19
|
Ibrahim WW, Sayed RH, Kandil EA, Wadie W. Niacin mitigates blood-brain barrier tight junctional proteins dysregulation and cerebral inflammation in ketamine rat model of psychosis: Role of GPR109A receptor. Prog Neuropsychopharmacol Biol Psychiatry 2022; 119:110583. [PMID: 35690118 DOI: 10.1016/j.pnpbp.2022.110583] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Revised: 06/05/2022] [Accepted: 06/05/2022] [Indexed: 01/25/2023]
Abstract
Dysregulated inflammatory responses and blood-brain barrier (BBB) dysfunction are recognized as central factors in the development of psychiatric disorders. The present study was designed to evaluate the effect of niacin on BBB integrity in ketamine-induced model of psychosis. Meanwhile, mepenzolate bromide (MPN), a GPR109A receptor blocker, was used to investigate the role of this receptor on the observed niacin's effect. Male Wistar rats received ketamine (30 mg/kg/day, i.p) for 5 consecutive days and then niacin (40 mg/kg/day, p.o), with or without MPN (5 mg/kg/day, i.p), was given for the subsequent 15 days. Three days before the end of experiment, rats were behaviorally tested using open field, novel object recognition, social interaction, and forced swimming tests. Niacin significantly ameliorated ketamine-induced behavioral deficits, amended gamma aminobutyric acid and glutamate concentration, decreased tumor necrosis factor-α and matrix metallopeptidase 9 levels, and increased netrin-1 contents in the hippocampus of rats. Niacin also augmented the hippocampal expression of ZO-1, occludin, and claudin-5 proteins, indicating the ability of niacin to restore the BBB integrity. Moreover, the histopathologic changes in hippocampal neurons were alleviated. Since all the beneficial effects of niacin in the present investigation were partially abolished by the co-administration of MPN; GPR109A receptor was proven to partially mediate the observed antipsychotic effects of niacin. These data revealed that GPR109A-mediated signaling pathways might represent potential targets for therapeutic interventions to prevent or slow the progression of psychosis.
Collapse
Affiliation(s)
- Weam W Ibrahim
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Egypt
| | - Rabab H Sayed
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Egypt.
| | - Esraa A Kandil
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Egypt
| | - Walaa Wadie
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Egypt
| |
Collapse
|
20
|
Central and peripheral regulations mediated by short-chain fatty acids on energy homeostasis. Transl Res 2022; 248:128-150. [PMID: 35688319 DOI: 10.1016/j.trsl.2022.06.003] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/03/2022] [Revised: 05/16/2022] [Accepted: 06/01/2022] [Indexed: 11/24/2022]
Abstract
The human gut microbiota influences obesity, insulin resistance, and the subsequent development of type 2 diabetes (T2D). The gut microbiota digests and ferments nutrients resulting in the production of short-chain fatty acids (SCFAs), which generate various beneficial metabolic effects on energy and glucose homeostasis. However, their roles in the central nervous system (CNS)-mediated outputs on the metabolism have only been minimally studied. Here, we explore what is known and future directions that may be worth exploring in this emerging area. Specifically, we searched studies or data in English by using PubMed, Google Scholar, and the Human Metabolome Database. Studies were filtered by time from 1978 to March 2022. As a result, 195 studies, 53 reviews, 1 website, and 1 book were included. One hundred and sixty-five of 195 studies describe the production and metabolism of SCFAs or the effects of SCFAs on energy homeostasis, glucose balance, and mental diseases through the gut-brain axis or directly by a central pathway. Thirty of 195 studies show that inappropriate metabolism and excessive of SCFAs are metabolically detrimental. Most studies suggest that SCFAs exert beneficial metabolic effects by acting as the energy substrate in the TCA cycle, regulating the hormones related to satiety regulation and insulin secretion, and modulating immune cells and microglia. These functions have been linked with AMPK signaling, GPCRs-dependent pathways, and inhibition of histone deacetylases (HDACs). However, the studies focusing on the central effects of SCFAs are still limited. The mechanisms by which central SCFAs regulate appetite, energy expenditure, and blood glucose during different physiological conditions warrant further investigation.
Collapse
|
21
|
Maus KD, Stephenson DJ, Ali AN, MacKnight HP, Huang HJ, Serrats J, Kim M, Diegelmann RF, Chalfant CE. Ceramide kinase regulates acute wound healing by suppressing 5-oxo-ETE biosynthesis and signaling via its receptor OXER1. J Lipid Res 2022; 63:100187. [PMID: 35219746 PMCID: PMC8980959 DOI: 10.1016/j.jlr.2022.100187] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Revised: 02/18/2022] [Accepted: 02/21/2022] [Indexed: 01/10/2023] Open
Abstract
The sphingolipid, ceramide-1-phosphate (C1P), has been shown to promote the inflammatory phase and inhibit the proliferation and remodeling stages of wound repair via direct interaction with group IVA cytosolic phospholipase A2, a regulator of eicosanoid biosynthesis that fine-tunes the behaviors of various cell types during wound healing. However, the anabolic enzyme responsible for the production of C1P that suppresses wound healing as well as bioactive eicosanoids and target receptors that drive enhanced wound remodeling have not been characterized. Herein, we determined that decreasing C1P activity via inhibitors or genetic ablation of the anabolic enzyme ceramide kinase (CERK) significantly enhanced wound healing phenotypes. Importantly, postwounding inhibition of CERK enhanced the closure rate of acute wounds, improved the quality of healing, and increased fibroblast migration via a "class switch" in the eicosanoid profile. This switch reduced pro-inflammatory prostaglandins (e.g., prostaglandin E2) and increased levels of 5-hydroxyeicosatetraenoic acid and the downstream metabolite 5-oxo-eicosatetraenoic acid (5-oxo-ETE). Moreover, dermal fibroblasts from mice with genetically ablated CERK showed enhanced wound healing markers, while blockage of the murine 5-oxo-ETE receptor (oxoeicosanoid receptor 1) inhibited the enhanced migration phenotype of these cell models. Together, these studies reinforce the vital roles eicosanoids play in the wound healing process and demonstrate a novel role for CERK-derived C1P as a negative regulator of 5-oxo-ETE biosynthesis and the activation of oxoeicosanoid receptor 1 in wound healing. These findings provide foundational preclinical results for the use of CERK inhibitors to shift the balance from inflammation to resolution and increase the wound healing rate.
Collapse
Affiliation(s)
- Kenneth D Maus
- Department of Cell Biology, Microbiology, and Molecular Biology, University of South Florida, Tampa, FL, USA
| | - Daniel J Stephenson
- Department of Cell Biology, Microbiology, and Molecular Biology, University of South Florida, Tampa, FL, USA
| | - Anika N Ali
- Department of Cell Biology, Microbiology, and Molecular Biology, University of South Florida, Tampa, FL, USA
| | - Henry Patrick MacKnight
- Department of Cell Biology, Microbiology, and Molecular Biology, University of South Florida, Tampa, FL, USA
| | - Huey-Jing Huang
- Neuroscience Drug Discovery Unit, Takeda California, San Diego, CA, USA
| | - Jordi Serrats
- Neuroscience Drug Discovery Unit, Takeda California, San Diego, CA, USA
| | - Minjung Kim
- Department of Cell Biology, Microbiology, and Molecular Biology, University of South Florida, Tampa, FL, USA
| | - Robert F Diegelmann
- Department of Biochemistry and Molecular Biology, Virginia Commonwealth University-School of Medicine, Richmond, VA, USA
| | - Charles E Chalfant
- Department of Cell Biology, Microbiology, and Molecular Biology, University of South Florida, Tampa, FL, USA; Cancer Biology and Evolution Program, The Moffitt Cancer Center, Tampa, FL, USA; Research Service, James A. Haley Veterans Hospital, Tampa, FL, USA; Division of Hematology & Oncology, Department of Medicine, University of Virginia, Charlottesville, VA, USA; Department of Cell Biology, University of Virginia, Charlottesville, VA, USA; Program in Cancer Biology, University of Virginia Cancer Center, Charlottesville, VA, USA; Research Service, Hunter Holmes McGuire Veterans Administration Medical Center, Richmond, VA, USA.
| |
Collapse
|
22
|
Strassheim D, Sullivan T, Irwin DC, Gerasimovskaya E, Lahm T, Klemm DJ, Dempsey EC, Stenmark KR, Karoor V. Metabolite G-Protein Coupled Receptors in Cardio-Metabolic Diseases. Cells 2021; 10:3347. [PMID: 34943862 PMCID: PMC8699532 DOI: 10.3390/cells10123347] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2021] [Revised: 11/10/2021] [Accepted: 11/18/2021] [Indexed: 12/15/2022] Open
Abstract
G protein-coupled receptors (GPCRs) have originally been described as a family of receptors activated by hormones, neurotransmitters, and other mediators. However, in recent years GPCRs have shown to bind endogenous metabolites, which serve functions other than as signaling mediators. These receptors respond to fatty acids, mono- and disaccharides, amino acids, or various intermediates and products of metabolism, including ketone bodies, lactate, succinate, or bile acids. Given that many of these metabolic processes are dysregulated under pathological conditions, including diabetes, dyslipidemia, and obesity, receptors of endogenous metabolites have also been recognized as potential drug targets to prevent and/or treat metabolic and cardiovascular diseases. This review describes G protein-coupled receptors activated by endogenous metabolites and summarizes their physiological, pathophysiological, and potential pharmacological roles.
Collapse
Affiliation(s)
- Derek Strassheim
- Department of Medicine Cardiovascular and Pulmonary Research Laboratory, University of Colorado Denver, Denver, CO 80204, USA; (D.S.); (T.S.); (D.C.I.); (E.G.); (D.J.K.); (E.C.D.); (K.R.S.)
| | - Timothy Sullivan
- Department of Medicine Cardiovascular and Pulmonary Research Laboratory, University of Colorado Denver, Denver, CO 80204, USA; (D.S.); (T.S.); (D.C.I.); (E.G.); (D.J.K.); (E.C.D.); (K.R.S.)
| | - David C. Irwin
- Department of Medicine Cardiovascular and Pulmonary Research Laboratory, University of Colorado Denver, Denver, CO 80204, USA; (D.S.); (T.S.); (D.C.I.); (E.G.); (D.J.K.); (E.C.D.); (K.R.S.)
| | - Evgenia Gerasimovskaya
- Department of Medicine Cardiovascular and Pulmonary Research Laboratory, University of Colorado Denver, Denver, CO 80204, USA; (D.S.); (T.S.); (D.C.I.); (E.G.); (D.J.K.); (E.C.D.); (K.R.S.)
| | - Tim Lahm
- Division of Pulmonary, Critical Care and Sleep Medicine, National Jewish Health Denver, Denver, CO 80206, USA;
- Rocky Mountain Regional VA Medical Center, Aurora, CO 80045, USA
| | - Dwight J. Klemm
- Department of Medicine Cardiovascular and Pulmonary Research Laboratory, University of Colorado Denver, Denver, CO 80204, USA; (D.S.); (T.S.); (D.C.I.); (E.G.); (D.J.K.); (E.C.D.); (K.R.S.)
- Rocky Mountain Regional VA Medical Center, Aurora, CO 80045, USA
- Division of Pulmonary Sciences and Critical Care Medicine, School of Medicine, University of Colorado, Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Edward C. Dempsey
- Department of Medicine Cardiovascular and Pulmonary Research Laboratory, University of Colorado Denver, Denver, CO 80204, USA; (D.S.); (T.S.); (D.C.I.); (E.G.); (D.J.K.); (E.C.D.); (K.R.S.)
- Rocky Mountain Regional VA Medical Center, Aurora, CO 80045, USA
- Division of Pulmonary Sciences and Critical Care Medicine, School of Medicine, University of Colorado, Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Kurt R. Stenmark
- Department of Medicine Cardiovascular and Pulmonary Research Laboratory, University of Colorado Denver, Denver, CO 80204, USA; (D.S.); (T.S.); (D.C.I.); (E.G.); (D.J.K.); (E.C.D.); (K.R.S.)
| | - Vijaya Karoor
- Department of Medicine Cardiovascular and Pulmonary Research Laboratory, University of Colorado Denver, Denver, CO 80204, USA; (D.S.); (T.S.); (D.C.I.); (E.G.); (D.J.K.); (E.C.D.); (K.R.S.)
- Division of Pulmonary, Critical Care and Sleep Medicine, National Jewish Health Denver, Denver, CO 80206, USA;
- Division of Pulmonary Sciences and Critical Care Medicine, School of Medicine, University of Colorado, Anschutz Medical Campus, Aurora, CO 80045, USA
| |
Collapse
|
23
|
Ghimire S, Weber D, Hippe K, Meedt E, Hoepting M, Kattner AS, Hiergeist A, Gessner A, Matos C, Ghimire S, Wolff D, Edinger M, Hoffmann P, Poeck H, Herr W, Holler E. GPR Expression in Intestinal Biopsies From SCT Patients Is Upregulated in GvHD and Is Suppressed by Broad-Spectrum Antibiotics. Front Immunol 2021; 12:753287. [PMID: 34777363 PMCID: PMC8588834 DOI: 10.3389/fimmu.2021.753287] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Accepted: 10/11/2021] [Indexed: 12/12/2022] Open
Abstract
Microbiota can exert immunomodulatory effects by short-chain fatty acids (SCFA) in experimental models of graft-versus-host disease (GvHD) after allogeneic hematopoietic stem cell transplantation (allo-SCT). Therefore we aimed to analyze the expression of SCFAs sensing G-protein coupled receptor GPR109A and GPR43 by quantitative PCR in 338 gastrointestinal (GI) biopsies obtained from 199 adult patients undergoing allo-SCT and assessed the interaction of GPR with FOXP3 expression and regulatory T cell infiltrates. GPR expression was strongly upregulated in patients with stage II-IV GvHD (p=0.000 for GPR109A, p=0.01 for GPR43) and at the onset of GvHD (p 0.000 for GPR109A, p=0.006 for GPR43) and correlated strongly with FOXP3 and NLRP3 expression. The use of broad-spectrum antibiotics (Abx) drastically suppressed GPR expression as well as FOXP3 expression in patients’ gut biopsies (p=0.000 for GPRs, FOXP3 mRNA and FOXP3+ cellular infiltrates). Logistic regression analysis revealed treatment with Abx as an independent factor associated with GPR and FOXP3 loss. The upregulation of GPRs was evident only in the absence of Abx (p=0.001 for GPR109A, p=0.014 for GPR43) at GvHD onset. Thus, GPR expression seems to be upregulated in the presence of commensal bacteria and associates with infiltration of FOXP3+ T regs, suggesting a protective, regenerative immunomodulatory response. However, Abx, which has been shown to induce dysbiosis, interferes with this protective response.
Collapse
Affiliation(s)
- Sakhila Ghimire
- Department of Internal Medicine III, University Hospital Regensburg, Regensburg, Germany
| | - Daniela Weber
- Department of Internal Medicine III, University Hospital Regensburg, Regensburg, Germany
| | - Katrin Hippe
- Department of Pathology, University of Regensburg, Regensburg, Germany
| | - Elisabeth Meedt
- Department of Internal Medicine III, University Hospital Regensburg, Regensburg, Germany
| | - Matthias Hoepting
- Department of Internal Medicine III, University Hospital Regensburg, Regensburg, Germany
| | - Anna-Sophia Kattner
- Department of Internal Medicine III, University Hospital Regensburg, Regensburg, Germany
| | - Andreas Hiergeist
- Institute for Medical Microbiology and Hygiene (IMHR), University Hospital Regensburg, Regensburg, Germany
| | - André Gessner
- Institute for Medical Microbiology and Hygiene (IMHR), University Hospital Regensburg, Regensburg, Germany
| | - Carina Matos
- Department of Internal Medicine III, University Hospital Regensburg, Regensburg, Germany
| | - Saroj Ghimire
- Kathmandu University School of Medical Sciences, Dhulikhel, Nepal
| | - Daniel Wolff
- Department of Internal Medicine III, University Hospital Regensburg, Regensburg, Germany
| | - Matthias Edinger
- Department of Internal Medicine III, University Hospital Regensburg, Regensburg, Germany.,Regensburg Center for Interventional Immunology (RCI), Regensburg, Germany
| | - Petra Hoffmann
- Department of Internal Medicine III, University Hospital Regensburg, Regensburg, Germany.,Regensburg Center for Interventional Immunology (RCI), Regensburg, Germany
| | - Hendrik Poeck
- Department of Internal Medicine III, University Hospital Regensburg, Regensburg, Germany
| | - Wolfgang Herr
- Department of Internal Medicine III, University Hospital Regensburg, Regensburg, Germany
| | - Ernst Holler
- Department of Internal Medicine III, University Hospital Regensburg, Regensburg, Germany
| |
Collapse
|
24
|
Song Y, Yang Y, Zeng W, Loor JJ, Jiang Q, Peng Z, Li Y, Jiang S, Feng X, Du X, Li X, Liu G. β-Hydroxybutyrate impairs neutrophil migration distance through activation of a protein kinase C and myosin light chain 2 signaling pathway in ketotic cows. J Dairy Sci 2021; 105:761-771. [PMID: 34635355 DOI: 10.3168/jds.2021-20875] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Accepted: 08/24/2021] [Indexed: 11/19/2022]
Abstract
Ketosis in dairy cows often occurs in the peripartal period and is accompanied by immune dysfunction. High concentrations of β-hydroxybutyrate (BHB) in peripheral blood during ketosis are closely related to the impairment of polymorphonuclear neutrophil (PMN) chemotaxis and contribute to immune dysfunction. The specific effect of BHB on PMN chemotaxis in dairy cows and the underlying molecular mechanisms are unclear. Here, 30 multiparous cows (within 3 wk postpartum) classified based on serum BHB as control (n = 15, BHB <0.6 mM) or clinically ketotic (n = 15, BHB >3.0 mM) were used. Blood samples were collected before feeding, and the isolated PMN were treated with platelet-activating factor for 0.5 h to activate their migration. Scanning electron microscopy revealed a longer tail in PMN of ketotic cows. In addition, the phosphorylation and transcription levels of myosin light chain 2 (MLC2) increased in PMN of ketotic cows. Polymorphonuclear neutrophils from control dairy cows were incubated with 3.0 mM BHB for different times in vitro, and 6 h was selected as the proper duration of BHB stimulation according to its inhibition effect on PMN migration using an under-agarose PMN chemotaxis model. Similarly, BHB stimulation in vitro resulted in inhibition of migration distance and deviation of migration direction of PMN, as well as a longer tail in morphology in the scanning electron microscope data, suggesting that BHB-induced PMN migration inhibition may be mediated by impairing the trailing edge contraction. To confirm this hypothesis, sotrastaurin (Sotra)-a specific inhibitor of protein kinase C (PKC), which is the core regulator of cell contraction-was used with or without BHB treatment in vitro. Sotra was pretreated 0.5 h before BHB treatment. Accordingly, BHB treatment increased the phosphorylation level of PKC and MLC2, the protein abundance of RhoA and rho-kinase 1 (ROCK1), and the mRNA abundance of PRKCA, MYL2, RHOA, and ROCK1 in PMN. In contrast, these effects of BHB on PMN were dampened by Sotra. As demonstrated by immunofluorescence experiments in vitro, the BHB-induced inhibition of trailing edge contraction of PMN was relieved by Sotra. In addition, Sotra also dampened the effects of BHB on PMN migration in vitro. Furthermore, as verified by in vivo experiments, compared with the control cows, both abundance and activation of PKC signaling were enhanced in PMN of ketotic cows. Overall, the present study revealed that high concentrations of blood BHB impaired PMN migration distance through inhibition of the trailing edge contraction, mediated by enhancing the activation of PKC-MLC2 signaling. These findings help explain the dysfunctional immune state in ketotic cows and provide information on the pathogenesis of infectious diseases secondary to ketosis.
Collapse
Affiliation(s)
- Yuxiang Song
- Key Laboratory of Zoonosis, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, Jilin Province, 130062, China
| | - Yuchen Yang
- Key Laboratory of Zoonosis, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, Jilin Province, 130062, China
| | - Wen Zeng
- Key Laboratory of Zoonosis, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, Jilin Province, 130062, China
| | - Juan J Loor
- Mammalian NutriPhysioGenomics, Department of Animal Sciences and Division of Nutritional Sciences, University of Illinois, Urbana 61801
| | - Qianming Jiang
- Mammalian NutriPhysioGenomics, Department of Animal Sciences and Division of Nutritional Sciences, University of Illinois, Urbana 61801
| | - Zhicheng Peng
- Key Laboratory of Zoonosis, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, Jilin Province, 130062, China
| | - Yunfei Li
- Key Laboratory of Zoonosis, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, Jilin Province, 130062, China
| | - Shang Jiang
- Key Laboratory of Zoonosis, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, Jilin Province, 130062, China
| | - Xiancheng Feng
- Key Laboratory of Zoonosis, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, Jilin Province, 130062, China
| | - Xiliang Du
- Key Laboratory of Zoonosis, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, Jilin Province, 130062, China
| | - Xinwei Li
- Key Laboratory of Zoonosis, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, Jilin Province, 130062, China.
| | - Guowen Liu
- Key Laboratory of Zoonosis, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, Jilin Province, 130062, China.
| |
Collapse
|
25
|
Chen H, Birnbaum Y, Ye R, Yang HC, Bajaj M, Ye Y. SGLT2 Inhibition by Dapagliflozin Attenuates Diabetic Ketoacidosis in Mice with Type-1 Diabetes. Cardiovasc Drugs Ther 2021; 36:1091-1108. [PMID: 34448973 DOI: 10.1007/s10557-021-07243-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 08/19/2021] [Indexed: 12/29/2022]
Abstract
BACKGROUND SGLT2 inhibitors increase plasma ketone concentrations. It has been suggested that insulinopenia, along with an increase in the counter-regulatory hormones epinephrine, corticosterone, glucagon and growth hormone, can induce ketoacidosis, especially in type-1 diabetes (T1DM). Dehydration precipitates SGLT2 inhibitor-induced ketoacidosis in type-2 diabetes. We studied the effects of dapagliflozin and water deprivation on the development of ketoacidosis and the associated signaling pathways in T1DM mice. METHODS C57BL/6 mice were fed a high-fat diet. After 7 days, some mice received intraperitoneal injection of streptozocin + alloxan (STZ/ALX). The treatment groups were control + water at lib; control + dapagloflozin + water at lib; control + dapagloflozin + water deprivation; STZ/ALX + water at lib; STZ/ALX + water deprivation; STZ/ALX + dapagloflozin + water at lib; STZ/ALX + dapagloflozin + water deprivation. Dapagliflozin was given for 7 days. In the morning of day 18, food was removed, and water was removed in the water deprivation groups. ELISA, rt-PCR, and immunoblotting were used to assess blood, heart, liver, white and brown adipose tissues. RESULTS The T1DM mice had ketoacidosis even without water deprivation. Water deprivation increased plasma levels of β-hydroxybutyrate, acetoacetate, corticosterone, and epinephrine and reduced the levels of adiponectin in T1DM mice. Interleukin (IL) 1β, IL-6, IL-8, and TNFα were also increased in the T1DM mice with water deprivation. Dapagliflozin attenuated the changes in the T1DM mice without and with water deprivation. Likewise, water deprivation increased the activation of the inflammasome in the heart, liver, and white fat of the T1DM mice and dapagliflozin attenuated these changes. Dapagliflozin reduced the mRNA levels of glucagon receptors in the liver and the increase in GPR109a in white and brown fat. In the liver, dapagliflozin increased AMPK phosphorylation, and attenuated the phosphorylation of TBK1 and the activation of NFκB. CONCLUSIONS Dapagliflozin reduced ketone body levels and attenuated the activation of NFκB and the activation of the inflammasome in T1DM mice with ketoacidosis.
Collapse
Affiliation(s)
- Huan Chen
- The Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, 301 University Blvd, BSB 648, Galveston, TX, 77555, USA.,Department of Acupuncture, First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Yochai Birnbaum
- The Section of Cardiology, Department of Medicine, Baylor College of Medicine, Houston, TX, USA
| | - Regina Ye
- The Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, 301 University Blvd, BSB 648, Galveston, TX, 77555, USA
| | - Hsiu-Chiung Yang
- Research and Early Development Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Göteborg, Sweden
| | - Mandeep Bajaj
- Section of Endocrinology, Baylor College of Medicine, Houston, TX, USA
| | - Yumei Ye
- The Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, 301 University Blvd, BSB 648, Galveston, TX, 77555, USA.
| |
Collapse
|
26
|
Swartz TH, Bradford BJ, Mamedova LK. Connecting Metabolism to Mastitis: Hyperketonemia Impaired Mammary Gland Defenses During a Streptococcus uberis Challenge in Dairy Cattle. Front Immunol 2021; 12:700278. [PMID: 34267762 PMCID: PMC8276125 DOI: 10.3389/fimmu.2021.700278] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Accepted: 06/15/2021] [Indexed: 11/17/2022] Open
Abstract
β-hydroxybutyrate (BHB) has been associated with disease incidence in early lactation dairy cattle, but such associations do not demonstrate causation. Therefore, the objective of this study was to examine the effects of BHB during an intramammary Streptococcus uberis challenge. A secondary objective was to elucidate the mechanisms behind BHB effects on cytokine transcript abundance using the RAW 264.7 cell line. Late lactation multiparous dairy cows (n = 12) were continuously infused intravenously with either BHB to induce hyperketonemia (target concentration: 1.8 mM) or with saline (CON) for 72 h during a S. uberis intramammary challenge. Body temperature, dry matter intake (DMI), milk production, and milk S. uberis cfu were measured daily until one week post-challenge. Blood samples were collected during infusion to assess changes in metabolism (glucose, insulin, glucagon, NEFA, and cortisol) and systemic inflammation (IL-1β and SAA). Mammary biopsies were conducted at 72 h post-challenge to assess transcript abundance of inflammation-associated genes. BHB-infused cows exhibited a delayed febrile response, noted by a lesser vaginal temperature during the final day of infusion, followed by a greater vaginal temperature 6 d post-challenge. Consequently, BHB-infused cows had greater S. uberis cfu on d 4, 6, and 7 as compared to CON. Accordingly, BHB-infused cows consumed less DM, produced less milk, had reduced blood glucose, and had increased cortisol concentrations, however, no effects were seen on other systemic parameters or transcript abundance of inflammation-related genes in mammary tissue. To elucidate mechanisms behind the impaired immune defenses, RAW 264.7 cells were transfected with a GPR109A siRNA for 24 h and then treated with or without 1.8 mM BHB and challenged or left unchallenged with S. uberis for an additional 3 h. Transfection with siRNA reduced Gpr109a by 75%. Although BHB treatment did not significantly increase Il10, GPR109A knockdown as compared to the scrambled control reduced Il10 by 90% in S. uberis challenged macrophages treated with BHB, suggesting that macrophage immune responses to S. uberis can be altered via a GPR109A-dependent mechanism. Taken together, these data suggest that BHB altered the immune response promoting tolerance toward S. uberis rather than resistance.
Collapse
Affiliation(s)
- Turner H. Swartz
- Department of Animal Science, Michigan State University, East Lansing, MI, United States
- Department of Animal Sciences and Industry, Kansas State University, Manhattan, KS, United States
| | - Barry J. Bradford
- Department of Animal Science, Michigan State University, East Lansing, MI, United States
- Department of Animal Sciences and Industry, Kansas State University, Manhattan, KS, United States
| | - Laman K. Mamedova
- Department of Animal Science, Michigan State University, East Lansing, MI, United States
- Department of Animal Sciences and Industry, Kansas State University, Manhattan, KS, United States
| |
Collapse
|
27
|
Pérez MJ, Baden P, Deleidi M. Progresses in both basic research and clinical trials of NAD+ in Parkinson's disease. Mech Ageing Dev 2021; 197:111499. [PMID: 33989633 DOI: 10.1016/j.mad.2021.111499] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Revised: 04/07/2021] [Accepted: 05/07/2021] [Indexed: 10/21/2022]
Abstract
The decline of nicotinamide adenine dinucleotide (NAD+) levels is a hallmark of aging in multiple organisms and tissues, including the human brain. Hence, agents that increase intracellular NAD+ could have beneficial effects in aging and age-related neurodegenerative diseases. Disturbances in NAD+ metabolism have also been observed in Parkinson's disease (PD), supporting a link between neuronal bioenergetics failure and disease pathogenesis. Here, we review emerging findings revealing key roles for NAD+ and related metabolites in experimental models of dopaminergic neurodegeneration and in PD patients. We discuss how increased NAD+ levels might ameliorate disease phenotypes by restoring neuronal mitochondrial energy metabolism, promoting cellular proteostasis, and modulating the immune system. Finally, we describe ongoing clinical trials targeting NAD+ in PD and highlight the need for further investigations to better delineate the association between NAD+, brain aging and disease, and optimal strategies for efficiently and safely raising NAD+ levels. A more comprehensive understanding of the basic mechanisms linking NAD+, energy metabolism, and PD, and of the impact of life-long NAD+ targeting strategies, are critical to inform future clinical applications.
Collapse
Affiliation(s)
- María José Pérez
- German Center for Neurodegenerative Diseases (DZNE), 72076, Tübingen, Germany; Center of Neurology, Hertie Institute for Clinical Brain Research, University of Tübingen, 72076, Tübingen, Germany
| | - Pascale Baden
- German Center for Neurodegenerative Diseases (DZNE), 72076, Tübingen, Germany; Center of Neurology, Hertie Institute for Clinical Brain Research, University of Tübingen, 72076, Tübingen, Germany
| | - Michela Deleidi
- German Center for Neurodegenerative Diseases (DZNE), 72076, Tübingen, Germany; Center of Neurology, Hertie Institute for Clinical Brain Research, University of Tübingen, 72076, Tübingen, Germany.
| |
Collapse
|
28
|
Zhao M, Wang Z, Yang M, Ding Y, Zhao M, Wu H, Zhang Y, Lu Q. The Roles of Orphan G Protein-Coupled Receptors in Autoimmune Diseases. Clin Rev Allergy Immunol 2021; 60:220-243. [PMID: 33411320 DOI: 10.1007/s12016-020-08829-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/16/2020] [Indexed: 12/26/2022]
Abstract
G protein-coupled receptors (GPCRs) constitute the largest family of plasma membrane receptors in nature and mediate the effects of a variety of extracellular signals, such as hormone, neurotransmitter, odor, and light signals. Due to their involvement in a broad range of physiological and pathological processes and their accessibility, GPCRs are widely used as pharmacological targets of treatment. Orphan G protein-coupled receptors (oGPCRs) are GPCRs for which no natural ligands have been found, and they not only play important roles in various physiological functions, such as sensory perception, reproduction, development, growth, metabolism, and responsiveness, but are also closely related to many major diseases, such as central nervous system (CNS) diseases, metabolic diseases, and cancer. Recently, many studies have reported that oGPCRs play increasingly important roles as key factors in the occurrence and progression of autoimmune diseases. Therefore, oGPCRs are likely to become potential therapeutic targets and may provide a breakthrough in the study of autoimmune diseases. In this article, we focus on reviewing the recent research progress and clinical treatment effects of oGPCRs in three common autoimmune diseases: multiple sclerosis (MS), rheumatoid arthritis (RA), and systemic lupus erythematosus (SLE), shedding light on novel strategies for treatments.
Collapse
Affiliation(s)
- Mingming Zhao
- Department of Dermatology, Hunan Key Laboratory of Medical Epigenomics, Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Zheyu Wang
- University of South China, Hengyang, Hunan, China.,Maternal & Child Health Care Hospital Hainan Province, Haikou, Hainan, China
| | - Ming Yang
- Department of Dermatology, Hunan Key Laboratory of Medical Epigenomics, Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Yan Ding
- Maternal & Child Health Care Hospital Hainan Province, Haikou, Hainan, China.,Hainan Province Dermatol Disease Hospital, Haikou, Hainan, China
| | - Ming Zhao
- Department of Dermatology, Hunan Key Laboratory of Medical Epigenomics, Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Haijing Wu
- Department of Dermatology, Hunan Key Laboratory of Medical Epigenomics, Second Xiangya Hospital, Central South University, Changsha, Hunan, China.
| | - Yan Zhang
- Department of Biophysics, and Department of Pathology of Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, 310058, China. .,Zhejiang Laboratory for Systems & Precison Medicine, Zhejiang University Medical Center, Hangzhou, 311121, China. .,Zhejiang Provincial Key Laboratory of Immunity and Inflammatory Diseases, Hangzhou, 310058, China. .,MOE Frontier Science Center for Brain Research and Brain-Machine Integration, Zhejiang University School of Medicine, Hangzhou, 310058, China.
| | - Qianjin Lu
- Department of Dermatology, Hunan Key Laboratory of Medical Epigenomics, Second Xiangya Hospital, Central South University, Changsha, Hunan, China. .,Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, China.
| |
Collapse
|
29
|
Chen JR, Zhao H, Wankhade UD, Chintapalli SV, Li C, Gai D, Shankar K, Zhan F, Lazarenko OP. GPR109A mediates the effects of hippuric acid on regulating osteoclastogenesis and bone resorption in mice. Commun Biol 2021; 4:53. [PMID: 33420329 PMCID: PMC7794563 DOI: 10.1038/s42003-020-01564-2] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2020] [Accepted: 11/26/2020] [Indexed: 12/26/2022] Open
Abstract
The G protein-coupled receptor 109 A (GPR109A) is robustly expressed in osteoclastic precursor macrophages. Previous studies suggested that GPR109A mediates effects of diet-derived phenolic acids such as hippuric acid (HA) and 3-(3-hydroxyphenyl) propionic acid (3-3-PPA) on promoting bone formation. However, the role of GPR109A in metabolic bone homeostasis and osteoclast differentiation has not been investigated. Using densitometric, bone histologic and molecular signaling analytic methods, we uncovered that bone mass and strength were significantly higher in tibia and spine of standard rodent diet weaned 4-week-old and 6-month-old GPR109A gene deletion (GPR109A-/-) mice, compared to their wild type controls. Osteoclast numbers in bone and in ex vivo bone marrow cell cultures were significantly decreased in GPR109A-/- mice compared to wild type controls. In accordance with these data, CTX-1 in bone marrow plasma and gene expression of bone resorption markers (TNFα, TRAP, Cathepsin K) were significantly decreased in GPR109A-/- mice, while on the other hand, P1NP was increased in serum from both male and female GPR109A-/- mice compared to their respective controls. GPR109A deletion led to suppressed Wnt/β-catenin signaling in osteoclast precursors to inhibit osteoclast differentiation and activity. Indeed, HA and 3-3-PPA substantially inhibited RANKL-induced GPR109A expression and Wnt/β-catenin signaling in osteoclast precursors and osteoclast differentiation. Resultantly, HA significantly inhibited bone resorption and increased bone mass in wild type mice, but had no additional effects on bone in GPR109A-/- mice compared with their respective untreated control mice. These results suggest an important role for GPR109A during osteoclast differentiation and bone resorption mediating effects of HA and 3-3-PPA on inhibiting bone resorption during skeletal development.
Collapse
Affiliation(s)
- Jin-Ran Chen
- Arkansas Children's Nutrition Center, Little Rock, AR, 72202, USA.
- Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, AR, 72202, USA.
| | - Haijun Zhao
- Arkansas Children's Nutrition Center, Little Rock, AR, 72202, USA
- Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, AR, 72202, USA
| | - Umesh D Wankhade
- Arkansas Children's Nutrition Center, Little Rock, AR, 72202, USA
- Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, AR, 72202, USA
| | - Sree V Chintapalli
- Arkansas Children's Nutrition Center, Little Rock, AR, 72202, USA
- Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, AR, 72202, USA
| | - Can Li
- Myeloma Center, University of Arkansas for Medical Sciences, Little Rock, AR, 72202, USA
| | - Dongzheng Gai
- Myeloma Center, University of Arkansas for Medical Sciences, Little Rock, AR, 72202, USA
| | - Kartik Shankar
- Arkansas Children's Nutrition Center, Little Rock, AR, 72202, USA
- Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, AR, 72202, USA
- Department of Pediatrics, Section of Nutrition, University of Colorado School of Medicine, Aurora, CO, 80045, USA
| | - Fenghuang Zhan
- Myeloma Center, University of Arkansas for Medical Sciences, Little Rock, AR, 72202, USA
| | - Oxana P Lazarenko
- Arkansas Children's Nutrition Center, Little Rock, AR, 72202, USA
- Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, AR, 72202, USA
| |
Collapse
|
30
|
B Vitamins and Their Role in Immune Regulation and Cancer. Nutrients 2020; 12:nu12113380. [PMID: 33158037 PMCID: PMC7693142 DOI: 10.3390/nu12113380] [Citation(s) in RCA: 159] [Impact Index Per Article: 31.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Revised: 10/27/2020] [Accepted: 11/02/2020] [Indexed: 12/13/2022] Open
Abstract
B group vitamins represent essential micronutrients for myriad metabolic and regulatory processes required for human health, serving as cofactors used by hundreds of enzymes that carry out essential functions such as energy metabolism, DNA and protein synthesis and other critical functions. B vitamins and their corresponding vitamers are universally essential for all cellular life forms, from bacteria to humans. Humans are unable to synthesize most B vitamins and are therefore dependent on their diet for these essential micronutrients. More recently, another source of B vitamins has been identified which is derived from portions of the 1013 bacterial cells inhabiting the gastrointestinal tract. Here we review the expanding literature examining the relationship between B vitamins and the immune system and diverse cancers. Evidence of B vitamin’s role in immune cell regulation has accumulated in recent years and may help to clarify the disparate findings of numerous studies attempting to link B vitamins to cancer development. Much work remains to be carried out to fully clarify these relationships as the complexity of B vitamins’ essential functions complicates an unequivocal assessment of their beneficial or detrimental effects in inflammation and cancers.
Collapse
|
31
|
Sato FT, Yap YA, Crisma AR, Portovedo M, Murata GM, Hirabara SM, Ribeiro WR, Marcantonio Ferreira C, Cruz MM, Pereira JNB, Payolla TB, Guima SES, Thomas AM, Setubal JC, Alonso-Vale MIC, Santos MF, Curi R, Marino E, Vinolo MAR. Tributyrin Attenuates Metabolic and Inflammatory Changes Associated with Obesity through a GPR109A-Dependent Mechanism. Cells 2020; 9:E2007. [PMID: 32882837 PMCID: PMC7563536 DOI: 10.3390/cells9092007] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Revised: 08/21/2020] [Accepted: 08/24/2020] [Indexed: 12/21/2022] Open
Abstract
Obesity is linked with altered microbial short-chain fatty acids (SCFAs), which are a signature of gut dysbiosis and inflammation. In the present study, we investigated whether tributyrin, a prodrug of the SCFA butyrate, could improve metabolic and inflammatory profiles in diet-induced obese mice. Mice fed a high-fat diet for eight weeks were treated with tributyrin or placebo for another six weeks. We show that obese mice treated with tributyrin had lower body weight gain and an improved insulin responsiveness and glucose metabolism, partly via reduced hepatic triglycerides content. Additionally, tributyrin induced an anti-inflammatory state in the adipose tissue by reduction of Il-1β and Tnf-a and increased Il-10, Tregs cells and M2-macrophages. Moreover, improvement in glucose metabolism and reduction of fat inflammatory states associated with tributyrin treatment were dependent on GPR109A activation. Our results indicate that exogenous targeting of SCFA butyrate attenuates metabolic and inflammatory dysfunction, highlighting a potentially novel approach to tackle obesity.
Collapse
Affiliation(s)
- Fabio Takeo Sato
- Department of Genetics, Evolution, Microbiology and Immunology, Institute of Biology, University of Campinas, Campinas 13083007, Brazil; (F.T.S.); (M.P.)
| | - Yu Anne Yap
- Department of Biochemistry, Biomedicine Discovery Institute, Monash University, Melbourne 3800, Australia;
| | - Amanda Rabello Crisma
- Department of Clinical Analyses, Federal University of Paraná, Curitiba 80060000, Brazil;
| | - Mariana Portovedo
- Department of Genetics, Evolution, Microbiology and Immunology, Institute of Biology, University of Campinas, Campinas 13083007, Brazil; (F.T.S.); (M.P.)
| | - Gilson Masahiro Murata
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of São Paulo, São Paulo 05508000, Brazil; (G.M.M.); (T.B.P.); (R.C.)
| | - Sandro Massao Hirabara
- Interdisciplinary Postgraduate Program in Health Science, Cruzeiro do Sul University, São Paulo 01506000, Brazil; (S.M.H.); (J.N.B.P.)
| | - Willian Rodrigues Ribeiro
- Department of Pharmaceutics Sciences, Institute of Environmental Chemistry and Pharmaceutical Sciences, Federal University of São Paulo, Diadema 09972270, Brazil; (W.R.R.); (C.M.F.)
| | - Caroline Marcantonio Ferreira
- Department of Pharmaceutics Sciences, Institute of Environmental Chemistry and Pharmaceutical Sciences, Federal University of São Paulo, Diadema 09972270, Brazil; (W.R.R.); (C.M.F.)
| | - Maysa Mariana Cruz
- Department of Biological Sciences, Institute of Environmental, Chemical and Pharmaceutical Sciences, Federal University of São Paulo, Diadema 09972270, Brazil; (M.M.C.); (M.I.C.A.-V.)
| | - Joice Naiara Bertaglia Pereira
- Interdisciplinary Postgraduate Program in Health Science, Cruzeiro do Sul University, São Paulo 01506000, Brazil; (S.M.H.); (J.N.B.P.)
| | - Tanyara Baliani Payolla
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of São Paulo, São Paulo 05508000, Brazil; (G.M.M.); (T.B.P.); (R.C.)
| | - Suzana Eiko Sato Guima
- Department of Biochemistry, Institute of Chemistry, University of São Paulo, São Paulo 05508000, Brazil; (S.E.S.G.); (A.M.T.); (J.C.S.)
| | - Andrew Maltez Thomas
- Department of Biochemistry, Institute of Chemistry, University of São Paulo, São Paulo 05508000, Brazil; (S.E.S.G.); (A.M.T.); (J.C.S.)
| | - João Carlos Setubal
- Department of Biochemistry, Institute of Chemistry, University of São Paulo, São Paulo 05508000, Brazil; (S.E.S.G.); (A.M.T.); (J.C.S.)
| | - Maria Isabel Cardoso Alonso-Vale
- Department of Biological Sciences, Institute of Environmental, Chemical and Pharmaceutical Sciences, Federal University of São Paulo, Diadema 09972270, Brazil; (M.M.C.); (M.I.C.A.-V.)
| | - Marinilce Fagundes Santos
- Department of Cell and Developmental Biology, Institute of Biomedical Sciences, University of São Paulo, São Paulo 05508000, Brazil;
| | - Rui Curi
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of São Paulo, São Paulo 05508000, Brazil; (G.M.M.); (T.B.P.); (R.C.)
- Interdisciplinary Postgraduate Program in Health Science, Cruzeiro do Sul University, São Paulo 01506000, Brazil; (S.M.H.); (J.N.B.P.)
- Butantan Institute, São Paulo 05503900, Brazil
| | - Eliana Marino
- Department of Biochemistry, Biomedicine Discovery Institute, Monash University, Melbourne 3800, Australia;
| | - Marco A. R. Vinolo
- Department of Genetics, Evolution, Microbiology and Immunology, Institute of Biology, University of Campinas, Campinas 13083007, Brazil; (F.T.S.); (M.P.)
- Experimental Medicine Research Cluster (EMRC), University of Campinas (UNICAMP), Campinas 13083007, Brazil
- Obesity and Comorbidities Research Center (OCRC), University of Campinas (UNICAMP), Campinas 13083864, Brazil
| |
Collapse
|
32
|
Koutnik AP, Poff AM, Ward NP, DeBlasi JM, Soliven MA, Romero MA, Roberson PA, Fox CD, Roberts MD, D'Agostino DP. Ketone Bodies Attenuate Wasting in Models of Atrophy. J Cachexia Sarcopenia Muscle 2020; 11:973-996. [PMID: 32239651 PMCID: PMC7432582 DOI: 10.1002/jcsm.12554] [Citation(s) in RCA: 61] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/18/2019] [Revised: 01/18/2020] [Accepted: 01/30/2020] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND Cancer Anorexia Cachexia Syndrome (CACS) is a distinct atrophy disease negatively influencing multiple aspects of clinical care and patient quality of life. Although it directly causes 20% of all cancer-related deaths, there are currently no model systems that encompass the entire multifaceted syndrome, nor are there any effective therapeutic treatments. METHODS A novel model of systemic metastasis was evaluated for the comprehensive CACS (metastasis, skeletal muscle and adipose tissue wasting, inflammation, anorexia, anemia, elevated protein breakdown, hypoalbuminemia, and metabolic derangement) in both males and females. Ex vivo skeletal muscle analysis was utilized to determine ubiquitin proteasome degradation pathway activation. A novel ketone diester (R/S 1,3-Butanediol Acetoacetate Diester) was assessed in multifaceted catabolic environments to determine anti-atrophy efficacy. RESULTS Here, we show that the VM-M3 mouse model of systemic metastasis demonstrates a novel, immunocompetent, logistically feasible, repeatable phenotype with progressive tumor growth, spontaneous metastatic spread, and the full multifaceted CACS with sex dimorphisms across tissue wasting. We also demonstrate that the ubiquitin proteasome degradation pathway was significantly upregulated in association with reduced insulin-like growth factor-1/insulin and increased FOXO3a activation, but not tumor necrosis factor-α-induced nuclear factor-kappa B activation, driving skeletal muscle atrophy. Additionally, we show that R/S 1,3-Butanediol Acetoacetate Diester administration shifted systemic metabolism, attenuated tumor burden indices, reduced atrophy/catabolism and mitigated comorbid symptoms in both CACS and cancer-independent atrophy environments. CONCLUSIONS Our findings suggest the ketone diester attenuates multifactorial CACS skeletal muscle atrophy and inflammation-induced catabolism, demonstrating anti-catabolic effects of ketone bodies in multifactorial atrophy.
Collapse
Affiliation(s)
- Andrew P. Koutnik
- Department of Molecular Pharmacology and PhysiologyMorsani College of Medicine, University of South FloridaTampaFLUSA
| | - Angela M. Poff
- Department of Molecular Pharmacology and PhysiologyMorsani College of Medicine, University of South FloridaTampaFLUSA
| | - Nathan P. Ward
- Department of Cancer PhysiologyMoffitt Cancer Center, H. Lee Moffitt Cancer Center and Research InstituteTampaFLUSA
| | - Janine M. DeBlasi
- Department of Molecular Pharmacology and PhysiologyMorsani College of Medicine, University of South FloridaTampaFLUSA
| | - Maricel A. Soliven
- Department of Molecular Pharmacology and PhysiologyMorsani College of Medicine, University of South FloridaTampaFLUSA
| | | | | | - Carl D. Fox
- School of KinesiologyAuburn UniversityAuburnALUSA
| | | | - Dominic P. D'Agostino
- Department of Molecular Pharmacology and PhysiologyMorsani College of Medicine, University of South FloridaTampaFLUSA
- Institute for Human and Machine CognitionOcalaFLUSA
| |
Collapse
|
33
|
Effects of Dietary Supplements on the Bioaccessibility of Se, Zn and Cd in Rice: Preliminary Observations from In Vitro Gastrointestinal Simulation Tests. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2020; 17:ijerph17144978. [PMID: 32664443 PMCID: PMC7399922 DOI: 10.3390/ijerph17144978] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Revised: 07/07/2020] [Accepted: 07/08/2020] [Indexed: 12/31/2022]
Abstract
Trace elements such as selenium (Se) and zinc (Zn) are essential elements in the human body, while cadmium (Cd) has no physiological function. A high proportion of people consume dietary supplements to enhance the performance of the body or alter the nutrient contents within the body. Therefore, this study was conducted to evaluate the interaction effects of several popular dietary supplements on the bioaccessibility of Se, Zn and Cd in rice with the hope of identifying dietary supplements that can increase rice Se and Zn bioaccessibility but decrease rice Cd bioaccessibility. The results from in vitro gastrointestinal simulation tests showed that the bioaccessibility of these elements in rice was in the order of Cd (52.07%) > Zn (36.63%) > Se (10.19%) during the gastric phase and Zn (26.82%) > Cd (18.72%) > Se (14.70%) during the intestinal phase. The bioaccessibility of Se during the intestinal phase was greater than that during the gastric phase, and the bioaccessibility of Zn and Cd were the opposite. The bioaccessibility of Se significantly increased in response to vitamin C (VC), vitamin E (VE), vitamin B6 (VB6) and vitamin B9 (VB9), especially VC, which also increased the bioaccessibility of Zn and decreased that of Cd. Procyanidins (OPC), methionine (Met) and coenzyme Q10 (Q10) significantly reduced the bioaccessibility of Se. These results suggest that the reasonable use of dietary supplements can effectively regulate the in vivo contents of trace elements, which provide valuable information for developing health interventions to address problems for specific people, especially selenium-deficient people.
Collapse
|
34
|
Motawi TK, Sadik NAH, Hamed MA, Ali SA, Khalil WKB, Ahmed YR. Potential therapeutic effects of antagonizing adenosine A 2A receptor, curcumin and niacin in rotenone-induced Parkinson's disease mice model. Mol Cell Biochem 2020; 465:89-102. [PMID: 31820278 DOI: 10.1007/s11010-019-03670-0] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Accepted: 11/30/2019] [Indexed: 01/04/2023]
Abstract
Parkinson's disease (PD) is the second common age-related neurodegenerative disease. It is characterized by control loss of voluntary movements control, resting tremor, postural instability, bradykinesia, and rigidity. The aim of the present work is to evaluate curcumin, niacin, dopaminergic and non-dopaminergic drugs in mice model of Parkinson's disease through behavioral, biochemical, genetic and histopathological observations. Mice treated with rotenone rerecorded significant increase in adenosine A2A receptor (A2AR) gene expression, α synuclein, acetylcholinesterase (AchE), malondialdehyde (MDA), angiotensin-II (Ang-II), c-reactive protein (CRP), interleukin-6 (IL-6), caspase-3 (Cas-3) and DNA fragmentation levels as compared with the control group. While, significant decrease in dopamine (DA), norepinephrine (NE), serotonin (5-HT), superoxide dismutase (SOD), reduced glutathione (GSH), ATP, succinate and lactate dehydrogenases (SDH &LDH) levels were detected. Treatment with curcumin, niacin, adenosine A2AR antagonist; ZM241385 and their combination enhanced the animals' behavior and restored all the selected parameters with variable degrees of improvement. The brain histopathological features of hippocampal and substantia nigra regions confirmed our results. In conclusion, the combination of curcumin, niacin and ZM241385 recorded the most potent treatment effect in Parkinsonism mice followed by ZM241385, as a single treatment. ZM241385 succeeded to antagonize adenosine A2A receptor by diminishing its gene expression and ameliorating all biochemical parameters under investigation. The newly investigated agent; ZM241385 has almost the same pattern of improvement as the classical drug; Sinemet®. This could shed the light to the need of detailed studies on ZM241385 for its possible role as a promising treatment against PD. Additionally, food supplements such as curcumin and niacin were effective in Parkinson's disease eradication.
Collapse
Affiliation(s)
- Tarek K Motawi
- Biochemistry Department, Faculty of Pharmacy, Cairo University, Cairo, Egypt
| | - Nermin A H Sadik
- Biochemistry Department, Faculty of Pharmacy, Cairo University, Cairo, Egypt
| | - Manal A Hamed
- Department of Therapeutic Chemistry, National Research Centre, El-Buhouth St., Dokki, Giza, 12622, Egypt.
| | - Sanaa A Ali
- Department of Therapeutic Chemistry, National Research Centre, El-Buhouth St., Dokki, Giza, 12622, Egypt
| | - Wagdy K B Khalil
- Cell Biology Department, National Research Centre, El-Buhouth St., Dokki, Giza, 12622, Egypt
| | - Yomna R Ahmed
- Department of Therapeutic Chemistry, National Research Centre, El-Buhouth St., Dokki, Giza, 12622, Egypt
| |
Collapse
|
35
|
Ma X, Luo X, Zhou S, Huang Y, Chen C, Huang C, Shen L, Zhang P, Liu C. Hydroxycarboxylic Acid Receptor 2 Is a Zika Virus Restriction Factor That Can Be Induced by Zika Virus Infection Through the IRE1-XBP1 Pathway. Front Cell Infect Microbiol 2020; 9:480. [PMID: 32039055 PMCID: PMC6990111 DOI: 10.3389/fcimb.2019.00480] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2019] [Accepted: 12/31/2019] [Indexed: 12/13/2022] Open
Abstract
Zika virus (ZIKV) is an emerging arthropod-borne virus and belongs to the Flaviviridae family. The infection of ZIKV has become the global health crisis because of its rapid spread and association with severe neurological disorders, including congenital microcephaly and Guillain-Barre Syndrome. To identify host factors contributing to ZIKV pathogenesis, transcriptomic landscape in ZIKV-infected cells was examined with mRNA microarray analysis and we observed that the expression of hydroxycarboxylic acid receptor 2 (HCAR2) could be significantly induced by ZIKV infection. By utilizing two IRE1 inhibitors and XBP1-specific shRNAs, we revealed that the up-regulation of HCAR2 expression induced by ZIKV was dependent on the IRE1-XBP1 pathway. Through the CRISPR/Cas9 system, we generated HCAR2-deficient cell clones in two cell types (human lung carcinoma epithelial A549 cell and human hepatoma Huh7.5 cell). We found that the depletion of HCAR2 significantly increased the replication level of ZIKV, including RNA levels, protein expression levels, and viral titers. In addition, our data demonstrated that the antiviral effect of HCAR2 was not involved in viral entry process and was not dependent on its antilipolytic effect on nicotinic acid/HCAR2-mediated signaling pathway. Taken together, our results indicated that HCAR2 could function as a restriction factor in control of ZIKV replication, potentially providing a novel molecular target for anti-ZIKV therapeutics.
Collapse
Affiliation(s)
- Xiaocao Ma
- Department of Immunology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China.,Key Laboratory of Tropical Disease Control, Ministry of Education, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Xin Luo
- Department of Immunology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China.,Key Laboratory of Tropical Disease Control, Ministry of Education, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Shili Zhou
- Department of Immunology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China.,Key Laboratory of Tropical Disease Control, Ministry of Education, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Yanxia Huang
- Department of Immunology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China.,Key Laboratory of Tropical Disease Control, Ministry of Education, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Cancan Chen
- Department of Pathology, First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Changbai Huang
- Department of Immunology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China.,Key Laboratory of Tropical Disease Control, Ministry of Education, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Li Shen
- Department of Immunology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China.,Key Laboratory of Tropical Disease Control, Ministry of Education, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Ping Zhang
- Department of Immunology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China.,Key Laboratory of Tropical Disease Control, Ministry of Education, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Chao Liu
- Department of Immunology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China.,Key Laboratory of Tropical Disease Control, Ministry of Education, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
36
|
A comprehensive mechanistic review insight into the effects of micronutrients on toll-like receptors functions. Pharmacol Res 2019; 152:104619. [PMID: 31887355 DOI: 10.1016/j.phrs.2019.104619] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/27/2019] [Revised: 11/23/2019] [Accepted: 12/26/2019] [Indexed: 12/29/2022]
Abstract
Toll-like receptors (TLRs) are the special proteins receptors for recognition of molecules related to the pathogens. In this way, TLRs and secreted cytokines as a result of TLRs activation are involved in the inflammation pathways. So far, in vivo and in vitro studies have demonstrated that micronutrients (vitamins & minerals) with a broad range of effects on body health, can regulate TLRs signaling pathways. Current review aimed at determining the possible mechanisms of micronutrient effects on TLRs functions. In the aspect of gene expression, micronutrients have inconsistent effects on mRNA level of TLRs which are dependent on time, dose and type of studied TLR. Also, some micronutrients affect gene expression of TLRs signaling mediators namely TLRs adaptors like Myeloid differentiation primary response 88 (MyD88). In the aspect of TLRs signaling pathways, nuclear factor-κB (NF-κB) is an important mediator which is regulated by micronutrients. Also, the regulatory effects of micronutrients on phosphorylation reactions may be effective in the activation/inactivation of TLRs signaling mediators. In addition, zinc can regulate TLRs signaling indirectly via the zinc finger proteins which have contradictory effects on TLRs cascade. In conclusion, the relationship between micronutrients and TLRs signaling is complicated and depends on some known internal, external and genetic factors like form of studied micronutrient, cell type, TLR agonist, dose and time of exposure, inflammation, apoptosis, cell cycle, and environmental factors. Some unknown factors may be effective in TLRs response and as a result additional mechanistic studies are needed to elucidate exact effect of micronutrients on TLRs signaling.
Collapse
|
37
|
An X, Bao Q, Di S, Zhao Y, Zhao S, Zhang H, Lian F, Tong X. The interaction between the gut Microbiota and herbal medicines. Biomed Pharmacother 2019; 118:109252. [DOI: 10.1016/j.biopha.2019.109252] [Citation(s) in RCA: 59] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2019] [Revised: 07/18/2019] [Accepted: 07/18/2019] [Indexed: 12/14/2022] Open
|
38
|
Giri B, Belanger K, Seamon M, Bradley E, Purohit S, Chong R, Morgan JC, Baban B, Wakade C. Niacin Ameliorates Neuro-Inflammation in Parkinson's Disease via GPR109A. Int J Mol Sci 2019; 20:ijms20184559. [PMID: 31540057 PMCID: PMC6770365 DOI: 10.3390/ijms20184559] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2019] [Revised: 09/06/2019] [Accepted: 09/10/2019] [Indexed: 12/17/2022] Open
Abstract
In this study, we used macrophage RAW264.7 cells to elucidate the molecular mechanism underlying the anti-inflammatory actions of niacin. Anti-inflammatory actions of niacin and a possible role of its receptor GPR109A have been studied previously. However, the precise molecular mechanism of niacin’s action in reducing inflammation through GPR109A is unknown. Here we observed that niacin reduced the translocation of phosphorylated nuclear kappa B (p-NF-κB) induced by lipopolysaccharide (LPS) in the nucleus of RAW264.7 cells. The reduction in the nuclear translocation in turn decreased the expression of pro-inflammatory cytokines IL-1β, IL-6 in RAW264.7 cells. We observed a decrease in the nuclear translocation of p-NF-κB and the expression of inflammatory cytokines after knockdown of GPR109A in RAW264.7 cells. Our results suggest that these molecular actions of niacin are mediated via its receptor GPR109A (also known as HCAR2) by controlling the translocation of p-NF-κB to the nucleus. Overall, our findings suggest that niacin treatment may have potential in reducing inflammation by targeting GPR109A.
Collapse
Affiliation(s)
- Banabihari Giri
- Charlie Norwood VA Medical Center, Augusta, GA 30904, USA.
- Department of Physical Therapy, Augusta University, Augusta, GA 30912, USA.
| | - Kasey Belanger
- Department of Physiology, Augusta University, Augusta, GA 30912, USA.
| | - Marissa Seamon
- Charlie Norwood VA Medical Center, Augusta, GA 30904, USA.
- Department of Neuroscience, Augusta University, Augusta, GA 30912, USA.
| | - Eric Bradley
- Edward Via College of Osteopathic Medicine, Greenville, SC 29303, USA.
| | - Sharad Purohit
- Charlie Norwood VA Medical Center, Augusta, GA 30904, USA.
- Department of Undergraduate Health Professionals, Augusta University, Augusta, GA 30912, USA.
| | - Raymond Chong
- Charlie Norwood VA Medical Center, Augusta, GA 30904, USA.
- Department of Interdisciplinary Health Sciences, Augusta University, Augusta, GA 30912, USA.
| | - John C Morgan
- Charlie Norwood VA Medical Center, Augusta, GA 30904, USA.
- Department of Neurology, Augusta University, Augusta, GA 30912, USA.
| | - Babak Baban
- Department of Oral Biology and Diagnostic Sciences, Augusta University, Augusta, GA 30912, USA.
| | - Chandramohan Wakade
- Charlie Norwood VA Medical Center, Augusta, GA 30904, USA.
- Department of Physical Therapy, Augusta University, Augusta, GA 30912, USA.
- Department of Neuroscience, Augusta University, Augusta, GA 30912, USA.
- Department of Neurology, Augusta University, Augusta, GA 30912, USA.
| |
Collapse
|
39
|
Kimura I, Ichimura A, Ohue-Kitano R, Igarashi M. Free Fatty Acid Receptors in Health and Disease. Physiol Rev 2019; 100:171-210. [PMID: 31487233 DOI: 10.1152/physrev.00041.2018] [Citation(s) in RCA: 583] [Impact Index Per Article: 97.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Fatty acids are metabolized and synthesized as energy substrates during biological responses. Long- and medium-chain fatty acids derived mainly from dietary triglycerides, and short-chain fatty acids (SCFAs) produced by gut microbial fermentation of the otherwise indigestible dietary fiber, constitute the major sources of free fatty acids (FFAs) in the metabolic network. Recently, increasing evidence indicates that FFAs serve not only as energy sources but also as natural ligands for a group of orphan G protein-coupled receptors (GPCRs) termed free fatty acid receptors (FFARs), essentially intertwining metabolism and immunity in multiple ways, such as via inflammation regulation and secretion of peptide hormones. To date, several FFARs that are activated by the FFAs of various chain lengths have been identified and characterized. In particular, FFAR1 (GPR40) and FFAR4 (GPR120) are activated by long-chain saturated and unsaturated fatty acids, while FFAR3 (GPR41) and FFAR2 (GPR43) are activated by SCFAs, mainly acetate, butyrate, and propionate. In this review, we discuss the recent reports on the key physiological functions of the FFAR-mediated signaling transduction pathways in the regulation of metabolism and immune responses. We also attempt to reveal future research opportunities for developing therapeutics for metabolic and immune disorders.
Collapse
Affiliation(s)
- Ikuo Kimura
- Department of Applied Biological Science, Graduate School of Agriculture, Tokyo University of Agriculture and Technology, Fuchu-shi, Tokyo, Japan; AMED-CREST, Japan Agency for Medical Research and Development, Chiyoda-ku, Tokyo, Japan; and Department of Biochemistry, Kyoto University Graduate School of Pharmaceutical Science, Sakyo, Kyoto, Japan
| | - Atsuhiko Ichimura
- Department of Applied Biological Science, Graduate School of Agriculture, Tokyo University of Agriculture and Technology, Fuchu-shi, Tokyo, Japan; AMED-CREST, Japan Agency for Medical Research and Development, Chiyoda-ku, Tokyo, Japan; and Department of Biochemistry, Kyoto University Graduate School of Pharmaceutical Science, Sakyo, Kyoto, Japan
| | - Ryuji Ohue-Kitano
- Department of Applied Biological Science, Graduate School of Agriculture, Tokyo University of Agriculture and Technology, Fuchu-shi, Tokyo, Japan; AMED-CREST, Japan Agency for Medical Research and Development, Chiyoda-ku, Tokyo, Japan; and Department of Biochemistry, Kyoto University Graduate School of Pharmaceutical Science, Sakyo, Kyoto, Japan
| | - Miki Igarashi
- Department of Applied Biological Science, Graduate School of Agriculture, Tokyo University of Agriculture and Technology, Fuchu-shi, Tokyo, Japan; AMED-CREST, Japan Agency for Medical Research and Development, Chiyoda-ku, Tokyo, Japan; and Department of Biochemistry, Kyoto University Graduate School of Pharmaceutical Science, Sakyo, Kyoto, Japan
| |
Collapse
|
40
|
Zhao H, Lazarenko OP, Chen JR. Hippuric acid and 3-(3-hydroxyphenyl) propionic acid inhibit murine osteoclastogenesis through RANKL-RANK independent pathway. J Cell Physiol 2019; 235:599-610. [PMID: 31271661 PMCID: PMC6852477 DOI: 10.1002/jcp.28998] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2018] [Accepted: 06/04/2019] [Indexed: 12/22/2022]
Abstract
Nutritional factors influence bone development. Previous studies demonstrated that bone mass significantly increased with suppressed bone resorption in early life of rats fed with AIN-93G semi-purified diets supplemented with 10% whole blueberry (BB) powder for 2 weeks. However, the effects of increased phenolic acids in animal serum due to this diet on bone and bone resorption were unclear. This in vitro and in ex vivo study examined the effects of phenolic hippuric acid (HA) and 3-(3-hydroxyphenyl) propionic acid (3-3-PPA) on osteoclastic cell differentiation and bone resorption. We cultured murine osteoclast (macrophage) cell line, RAW 264.7 cells, and hematopoietic osteoclast progenitor cells (isolated from 4-week-old C57BL6/J mice) with 50 ng/ml of receptor activator of nuclear factor κ-Β ligand (RANKL). Morphologic studies showed decreased osteoclast number with treatment of 2.5% mouse serum from BB diet-fed animals compared with those treated with serum from standard casein diet-fed mice in both RAW 264.7 cell and primary cell cultures. HA and 3-3-PPA, but not 3-4-PPA, had dose-dependent suppressive effects on osteoclastogenesis and osteoclast resorptive activity in Corning osteo-assay plates. Signaling pathway analysis showed that after pretreatment with HA or 3-3-PPA, RANKL-stimulated increase of osteoclastogenic markers, such as nuclear factor of activated T-cells, cytoplasmic 1 and matrix metallopeptidase 9 gene/protein expression were blunted. Inhibitory effects of HA and 3-3-PPA on osteoclastogenesis utilized RANKL/RANK independent mediators. The study revealed that HA and 3-3-PPA significantly inhibited osteoclastogenesis and bone osteoclastic resorptive activity.
Collapse
Affiliation(s)
- Haijun Zhao
- Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, Arkansas.,Arkansas Children's Nutrition Center, Little Rock, Arkansas
| | - Oxana P Lazarenko
- Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, Arkansas.,Arkansas Children's Nutrition Center, Little Rock, Arkansas
| | - Jin-Ran Chen
- Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, Arkansas.,Arkansas Children's Nutrition Center, Little Rock, Arkansas
| |
Collapse
|
41
|
Scognamiglio M, Costa D, Sorriento A, Napoli C. Current Drugs and Nutraceuticals for the Treatment of Patients with Dyslipidemias. Curr Pharm Des 2019; 25:85-95. [DOI: 10.2174/1381612825666190130101108] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2018] [Accepted: 01/20/2019] [Indexed: 02/05/2023]
Abstract
Coronary heart disease (CHD) remains the leading cause of disability and death in industrialized Countries.
Among many conditions, which contribute to the etiology and progression of CHD, the presence of high
low density lipoprotein-cholesterol (LDL-C) levels represents the major risk factor. Therefore, the reduction of
LDL-C levels plays a key role in the management of patients with high or very high cardiovascular risk. Although
statins represent the gold standard therapy for the reduction of cholesterol levels, these drugs do not allow to
achieve target levels of LDL-C in all patients. Indeed, a significant number of patients resulted intolerants, especially
when the dosage increased. The availability of new lipid-lowering drugs, such as ezetimibe and PCSK9
inhibitors, may represent an important alternative or complement to the conventional lipid-lowering therapies.
However, long-term studies are still needed to define both efficacy and safety of use of these latter new drugs.
Some nutraceuticals may become an adequate and effective support in the management of some patients. To date,
several nutraceuticals with different mechanism of actions that provide a good tolerability are available as lipidlowering
agents. In particular, the most investigated are red yeast rice, phytosterols, berberine, beta-glucans and
soy. The aim of this review was to report recent data on the efficacy and safety of principle hypocholesterolemic
drugs available and to evaluate the possible role of some nutraceuticals as support therapy in the management of
patients with dyslipidemias.
Collapse
Affiliation(s)
- Michele Scognamiglio
- U.O.C. Division of Clinical Immunology, Immunohematology, Transfusion Medicine and Transplant Immunology, Clinical Department of Internal Medicine and Specialistics, Department of Medical, Surgical, Neurological, Metabolic and Geriatric Sciences, University of Campania , Italy
| | - Dario Costa
- U.O.C. Division of Clinical Immunology, Immunohematology, Transfusion Medicine and Transplant Immunology, Clinical Department of Internal Medicine and Specialistics, Department of Medical, Surgical, Neurological, Metabolic and Geriatric Sciences, University of Campania , Italy
| | - Antonio Sorriento
- U.O.C. Division of Clinical Immunology, Immunohematology, Transfusion Medicine and Transplant Immunology, Clinical Department of Internal Medicine and Specialistics, Department of Medical, Surgical, Neurological, Metabolic and Geriatric Sciences, University of Campania , Italy
| | - Claudio Napoli
- U.O.C. Division of Clinical Immunology, Immunohematology, Transfusion Medicine and Transplant Immunology, Clinical Department of Internal Medicine and Specialistics, Department of Medical, Surgical, Neurological, Metabolic and Geriatric Sciences, University of Campania , Italy
| |
Collapse
|
42
|
Parada Venegas D, De la Fuente MK, Landskron G, González MJ, Quera R, Dijkstra G, Harmsen HJM, Faber KN, Hermoso MA. Short Chain Fatty Acids (SCFAs)-Mediated Gut Epithelial and Immune Regulation and Its Relevance for Inflammatory Bowel Diseases. Front Immunol 2019; 10:277. [PMID: 30915065 PMCID: PMC6421268 DOI: 10.3389/fimmu.2019.00277] [Citation(s) in RCA: 2083] [Impact Index Per Article: 347.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2018] [Accepted: 01/31/2019] [Indexed: 12/13/2022] Open
Abstract
Ulcerative colitis (UC) and Crohn's disease (CD), collectively known as Inflammatory Bowel Diseases (IBD), are caused by a complex interplay between genetic, immunologic, microbial and environmental factors. Dysbiosis of the gut microbiome is increasingly considered to be causatively related to IBD and is strongly affected by components of a Western life style. Bacteria that ferment fibers and produce short chain fatty acids (SCFAs) are typically reduced in mucosa and feces of patients with IBD, as compared to healthy individuals. SCFAs, such as acetate, propionate and butyrate, are important metabolites in maintaining intestinal homeostasis. Several studies have indeed shown that fecal SCFAs levels are reduced in active IBD. SCFAs are an important fuel for intestinal epithelial cells and are known to strengthen the gut barrier function. Recent findings, however, show that SCFAs, and in particular butyrate, also have important immunomodulatory functions. Absorption of SCFAs is facilitated by substrate transporters like MCT1 and SMCT1 to promote cellular metabolism. Moreover, SCFAs may signal through cell surface G-protein coupled receptors (GPCRs), like GPR41, GPR43, and GPR109A, to activate signaling cascades that control immune functions. Transgenic mouse models support the key role of these GPCRs in controlling intestinal inflammation. Here, we present an overview of microbial SCFAs production and their effects on the intestinal mucosa with specific emphasis on their relevance for IBD. Moreover, we discuss the therapeutic potential of SCFAs for IBD, either applied directly or by stimulating SCFAs-producing bacteria through pre- or probiotic approaches.
Collapse
Affiliation(s)
- Daniela Parada Venegas
- Laboratory of Innate Immunity, Program of Immunology, Institute of Biomedical Sciences, Faculty of Medicine, Universidad de Chile, Santiago, Chile.,Department of Gastroenterology and Hepatology, University of Groningen, University Medical Center Groningen, Groningen, Netherlands
| | - Marjorie K De la Fuente
- Laboratory of Innate Immunity, Program of Immunology, Institute of Biomedical Sciences, Faculty of Medicine, Universidad de Chile, Santiago, Chile
| | - Glauben Landskron
- Laboratory of Innate Immunity, Program of Immunology, Institute of Biomedical Sciences, Faculty of Medicine, Universidad de Chile, Santiago, Chile
| | - María Julieta González
- Program of Cell and Molecular Biology, Faculty of Medicine, Institute of Biomedical Sciences, Universidad de Chile, Santiago, Chile
| | - Rodrigo Quera
- Inflammatory Bowel Diseases Program, Department of Gastroenterology, Clínica Las Condes, Santiago, Chile
| | - Gerard Dijkstra
- Department of Gastroenterology and Hepatology, University of Groningen, University Medical Center Groningen, Groningen, Netherlands
| | - Hermie J M Harmsen
- Department of Medical Microbiology, University of Groningen, University Medical Center Groningen, Groningen, Netherlands
| | - Klaas Nico Faber
- Department of Gastroenterology and Hepatology, University of Groningen, University Medical Center Groningen, Groningen, Netherlands.,Department of Laboratory Medicine, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Marcela A Hermoso
- Laboratory of Innate Immunity, Program of Immunology, Institute of Biomedical Sciences, Faculty of Medicine, Universidad de Chile, Santiago, Chile
| |
Collapse
|
43
|
Gut microbiota: a potential manipulator for host adipose tissue and energy metabolism. J Nutr Biochem 2019; 64:206-217. [PMID: 30553096 DOI: 10.1016/j.jnutbio.2018.10.020] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2018] [Revised: 07/30/2018] [Accepted: 10/28/2018] [Indexed: 12/14/2022]
|
44
|
Mandrika I, Tilgase A, Petrovska R, Klovins J. Hydroxycarboxylic Acid Receptor Ligands Modulate Proinflammatory Cytokine Expression in Human Macrophages and Adipocytes without Affecting Adipose Differentiation. Biol Pharm Bull 2019; 41:1574-1580. [PMID: 30270326 DOI: 10.1248/bpb.b18-00301] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Members of the hydroxycarboxylic acid receptor (HCA1-3) family are mainly expressed in adipocytes and immune cells. HCA2 ligand, niacin, has been used for decades as lipid-modifying drug. Recent studies suggest that HCA ligands can be involved in the modulation of inflammatory processes. In this study, we evaluated the effects of HCA1-3 ligands on adipose differentiation and cytokine expression in human adipocytes and macrophages. Simpson-Golabi-Behmel syndrome (SGBS) preadipocytes were induced to differentiate into adipocytes for 8 d in the presence or absence of HCA ligands and evaluated for lipid accumulation and adipogenic gene expression. The inhibitory effects of the ligands on the expression and production of cytokines were measured in lipopolysaccharide (LPS)-stimulated adipocytes and THP-1 macrophage cells. Preadipocytes treated with HCA ligands showed no changes in the capacity to differentiate into adipocytes and no significant alteration in peroxisome proliferator activated receptor γ (PPARγ) or its target gene expression. HCA2-3 ligands significantly downregulated LPS-induced expression of interleukin (IL)-6 (53-64%), tumor necrosis factor-α (TNF-α) (55-69%) and IL-8 (51-59%) in adipocytes and macrophages. IL-1β inhibition (58-68%) by HCA2-3 ligands was observed only in adipocytes. Furthermore, LPS increased the expression of HCA2-3 in adipocytes and macrophages and this expression was decreased by treatment with their ligands. These results suggest that HCA ligands modulated LPS-mediated pro-inflammatory gene expression in both macrophages and adipocytes without affecting adipogenesis. Therefore, targeting HCA2 and HCA3 would be beneficial in treating inflammation conditions associated with atherosclerosis and obesity.
Collapse
|
45
|
Bhargava P, Fitzgerald KC, Venkata SLV, Smith MD, Kornberg MD, Mowry EM, Haughey NJ, Calabresi PA. Dimethyl fumarate treatment induces lipid metabolism alterations that are linked to immunological changes. Ann Clin Transl Neurol 2018; 6:33-45. [PMID: 30656182 PMCID: PMC6331509 DOI: 10.1002/acn3.676] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2018] [Revised: 09/24/2018] [Accepted: 09/28/2018] [Indexed: 12/22/2022] Open
Abstract
Objective Identify metabolic changes produced by dimethyl fumarate (DMF) treatment and link them to immunological effects. Methods We enrolled 18 MS patients and obtained blood prior to DMF and 6 months postinitiation. We also enrolled 18 healthy controls for comparison. We performed global metabolomics on plasma and used weighted correlation network analysis (WGCNA) to identify modules of correlated metabolites. We identified modules that changed with treatment, followed by targeted metabolomics to corroborate changes identified in global analyses. We correlated changes in metabolite modules and individual metabolites with changes in immunological parameters. Results We identified alterations in lipid metabolism after DMF treatment – increases in two modules (phospholipids, lysophospholipids and plasmalogens) and reduction in one module (saturated and poly‐unsaturated fatty acids) eigen‐metabolite values (all P < 0.05). Change in the fatty acid module was greater in participants who developed lymphopenia and was strongly associated with both reduction in absolute lymphocyte counts (r = 0.65; P = 0.005) and change in CD8+ T cell subsets. We also noted significant correlation of change in lymphocyte counts with multiple fatty acid levels (measured by targeted or untargeted methods). Interpretation This study demonstrates that DMF treatment alters lipid metabolism and that changes in fatty acid levels are related to DMF‐induced immunological changes.
Collapse
Affiliation(s)
- Pavan Bhargava
- Department of Neurology Johns Hopkins University School of Medicine Baltimore Maryland
| | - Kathryn C Fitzgerald
- Department of Neurology Johns Hopkins University School of Medicine Baltimore Maryland
| | - Swarajya L V Venkata
- Department of Neurology Johns Hopkins University School of Medicine Baltimore Maryland
| | - Matthew D Smith
- Department of Neurology Johns Hopkins University School of Medicine Baltimore Maryland
| | - Michael D Kornberg
- Department of Neurology Johns Hopkins University School of Medicine Baltimore Maryland
| | - Ellen M Mowry
- Department of Neurology Johns Hopkins University School of Medicine Baltimore Maryland
| | - Norman J Haughey
- Department of Neurology Johns Hopkins University School of Medicine Baltimore Maryland
| | - Peter A Calabresi
- Department of Neurology Johns Hopkins University School of Medicine Baltimore Maryland
| |
Collapse
|
46
|
Ruscica M, Ferri N, Macchi C, Corsini A, Sirtori CR. Lipid lowering drugs and inflammatory changes: an impact on cardiovascular outcomes? Ann Med 2018; 50:461-484. [PMID: 29976096 DOI: 10.1080/07853890.2018.1498118] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Inflammatory changes are responsible for maintenance of the atherosclerotic process and may underlie some of the most feared vascular complications. Among the multiple mechanisms of inflammation, the arterial deposition of lipids and particularly of cholesterol crystals is the one responsible for the activation of inflammasome NLRP3, followed by the rise of circulating markers, mainly C-reactive protein (CRP). Elevation of lipoproteins, LDL but also VLDL and remnants, associates with increased inflammatory changes and coronary risk. Lipid lowering medications can reduce cholesterolemia and CRP: patients with elevations of both are at greatest cardiovascular (CV) risk and receive maximum benefit from therapy. Evaluation of the major drug series indicates that statins exert the largest LDL and CRP reduction, accompanied by reduced CV events. Other drugs, mainly active on the triglyceride/HDL axis, for example, PPAR agonists, may improve CRP and the lipid pattern, especially in patients with metabolic syndrome. PCSK9 antagonists, the newest most potent medications, do not induce significant changes in inflammatory markers, but patients with the highest baseline CRP levels show the best CV risk reduction. Parallel evaluation of lipids and inflammatory changes clearly indicates a significant link, both guiding to patients at highest risk, and to the best pharmacological approach. Key messages Lipid lowering agents with "pleiotropic" effects provide a more effective approach to CV prevention In CANTOS study, patients achieving on-treatment hsCRP concentrations ≤2 mg/L had a higher benefit in terms of reduction in major CV events The anti-inflammatory activity of PCSK9 antagonists appears to be of a minimal extent.
Collapse
Affiliation(s)
- M Ruscica
- a Dipartimento di Scienze Farmacologiche e Biomolecolari , Università degli Studi di Milano , Milan , Italy
| | - N Ferri
- b Dipartimento di Scienze del Farmaco , Università degli Studi di Padova , Padova , Italy
| | - C Macchi
- a Dipartimento di Scienze Farmacologiche e Biomolecolari , Università degli Studi di Milano , Milan , Italy
| | - A Corsini
- a Dipartimento di Scienze Farmacologiche e Biomolecolari , Università degli Studi di Milano , Milan , Italy
| | - C R Sirtori
- c Centro Dislipidemie , A.S.S.T. Grande Ospedale Metropolitano Niguarda , Milan , Italy
| |
Collapse
|
47
|
Wakade C, Giri B, Malik A, Khodadadi H, Morgan JC, Chong RK, Baban B. Niacin modulates macrophage polarization in Parkinson's disease. J Neuroimmunol 2018; 320:76-79. [DOI: 10.1016/j.jneuroim.2018.05.002] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2018] [Revised: 04/17/2018] [Accepted: 05/03/2018] [Indexed: 12/21/2022]
|
48
|
Adiels M, Chapman MJ, Robillard P, Krempf M, Laville M, Borén J. Niacin action in the atherogenic mixed dyslipidemia of metabolic syndrome: Insights from metabolic biomarker profiling and network analysis. J Clin Lipidol 2018; 12:810-821.e1. [DOI: 10.1016/j.jacl.2018.03.083] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2017] [Revised: 03/19/2018] [Accepted: 03/22/2018] [Indexed: 01/26/2023]
|
49
|
Dimethyl fumarate downregulates the immune response through the HCA 2/GPR109A pathway: Implications for the treatment of multiple sclerosis. Mult Scler Relat Disord 2018; 23:46-50. [PMID: 29763776 DOI: 10.1016/j.msard.2018.04.016] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2017] [Revised: 03/02/2018] [Accepted: 04/21/2018] [Indexed: 12/13/2022]
Abstract
BACKGROUND The mechanisms of action of dimethyl fumarate (DMF), and its metabolite, monomethyl fumarate (MMF), for the treatment of multiple sclerosis are not completely elucidated. OBJECTIVES To discuss the role of DMF/MMF-induced hydroxycarboxylic acid receptor 2 (HCA2/GPR109A) pathway activation in the immune response and treatment of MS. METHODS A narrative (traditional) review of the current literature. RESULTS Studies have shown that binding of DMF/MMF to HCA2 on dendritic cells inhibits the production of pro-inflammatory cytokines in vitro and in MS murine models. Evidence suggests that activation of HCA2 expressed in immune cells and gut epithelial cells by DMF/MMF, may induce anti-inflammatory responses in the intestinal mucosa. CONCLUSION Although the DMF/MMF mechanism of action remains unclear, evidence suggests that the activation of HCA2/GPR109A pathway downregulates the immune response and may activate anti-inflammatory response in the intestinal mucosa, possibly leading to reduction in CNS tissue damage in MS patients.
Collapse
|
50
|
Liang Y, Lin C, Zhang Y, Deng Y, Liu C, Yang Q. Probiotic mixture of Lactobacillus and Bifidobacterium alleviates systemic adiposity and inflammation in non-alcoholic fatty liver disease rats through Gpr109a and the commensal metabolite butyrate. Inflammopharmacology 2018; 26:1051-1055. [PMID: 29633106 DOI: 10.1007/s10787-018-0479-8] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2018] [Accepted: 03/30/2018] [Indexed: 01/01/2023]
Abstract
AIMS The study explored the systemic adiposity and inflammation through Gpr109a and the commensal metabolite butyrate during the treatment of non-alcoholic fatty liver disease rats with the probiotic mixture of Lactobacillus and Bifidobacterium for 16 weeks. METHODS Fifteen male SD rats were randomly divided into three groups of five rats each: normal control group (basal feed), high-fat diet (HFD) feeding group (83% basal feed + 10% lard oil + 5% sucrose + 1.5% cholesterol + 0.5% cholate), and probiotic mixture intervention group (HFD + 0.6 g kg-1 day-1 probiotic mixture). Body composition, serum lipids, serum inflammatory markers, Gpr109a, and the commensal metabolite butyrate were assessed. RESULTS Compared with HFD group, probiotic mixture significantly reduced body weight and the levels of serum FFA, TG, ALT, IL-1β, and IL-18 (P < 0.05). The levels of Gpr109a and the commensal metabolite butyrate also changed significantly (P < 0.05). CONCLUSIONS Probiotic mixture might inhibit systemic adiposity and inflammation through Gpr109a and the commensal metabolite butyrate in response to the insult of HFD.
Collapse
Affiliation(s)
- Yinji Liang
- School of Nursing, Jinan University, 601 Huangpu Road West, Guangzhou, 510632, Guangdong, China
| | - Chenli Lin
- School of Basic Medicine, Jinan University, 601 Huangpu Road West, Guangzhou, 510632, Guangdong, China
| | - Yupei Zhang
- School of Traditional Chinese Medicine, Jinan University, 601 Huangpu Road West, Guangzhou, 510632, Guangdong, China
| | - Yuanjun Deng
- School of Traditional Chinese Medicine, Jinan University, 601 Huangpu Road West, Guangzhou, 510632, Guangdong, China
| | - Chan Liu
- School of Basic Medicine, Jinan University, 601 Huangpu Road West, Guangzhou, 510632, Guangdong, China
| | - Qinhe Yang
- School of Traditional Chinese Medicine, Jinan University, 601 Huangpu Road West, Guangzhou, 510632, Guangdong, China.
| |
Collapse
|