1
|
Chen S, Dan L, Xiang L, He Q, Hu D, Gao Y. The role of gut flora-driven Th cell responses in preclinical rheumatoid arthritis. J Autoimmun 2025; 154:103426. [PMID: 40300482 DOI: 10.1016/j.jaut.2025.103426] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Revised: 03/24/2025] [Accepted: 04/22/2025] [Indexed: 05/01/2025]
Abstract
Rheumatoid arthritis (RA) is a chronic autoimmune inflammatory disorder with an immune pathogenesis that evolves over decades. Preclinical RA (PreRA) represents a dynamic immune phase preceding clinical RA, marked by the loss of autoimmune tolerance, the appearance of tissue-invasive effector T cells, and the production of autoantibodies (such as antibodies against citrullinated proteins and rheumatoid factors). Extensive research has demonstrated that gut microbiota influence mucosal T-cell responses, driving the progression of PreRA through multiple mechanisms, including altered intestinal permeability, gene-environment interactions, bacterial antigenic specificity, molecular mimicry, and metabolite production. Environmental risk factors such as smoking, hormonal changes, and high-sodium (Na) diets, may contribute to RA pathogenesis via the gut microbiome. The next challenge in RA research lies in developing therapeutic strategies to intervene during the asymptomatic autoimmune phase, where dietary adjustments, natural compounds, probiotics, and other approaches could effectively modulate gut flora to prevent or delay RA onset.
Collapse
Affiliation(s)
- Shuanglan Chen
- Department of Rheumatology and Immunology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610072, China
| | - Lijuan Dan
- Department of Infection, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610072, China
| | - Li Xiang
- Department of Rheumatology and Immunology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610072, China
| | - Qingman He
- Department of Rheumatology and Immunology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610072, China
| | - Dongsen Hu
- Sichuan Jinxin Xi'nan Women's and Children's Hospital Co., Ltd, Chengdu, 610023, China
| | - Yongxiang Gao
- Department of Rheumatology and Immunology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610072, China.
| |
Collapse
|
2
|
Cao W, Zhu S, Wang W, Wang Y, Yang S, Sun H. Microarc oxidation-PBAT composite coating on EK30 biodegradable magnesium alloys to enhance corrosion resistance and cytocompatibility. Colloids Surf B Biointerfaces 2025; 253:114720. [PMID: 40262305 DOI: 10.1016/j.colsurfb.2025.114720] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2025] [Revised: 04/13/2025] [Accepted: 04/16/2025] [Indexed: 04/24/2025]
Abstract
The rapid degradation rate of biodegradable magnesium alloys restricts their use in medical implants. Therefore, designing a protective coating with a slow degradation rate and good biocompatibility is crucial. For biodegradable magnesium alloy stents, it is equally important to enhance the material's corrosion resistance and ensure the coating's deformation adaptability to the stent. This study, utilized a combination of micro-arc oxidation (MAO) and dip-coating techniques to develop a novel composite coating comprising an MAO base layer and a poly(butylene adipate-co-terephthalate) (PBAT) outer layer on EK30 magnesium alloy. This composite coating was designed to enhance the corrosion resistance and biocompatibility of EK30 magnesium alloy for stent applications. The surface characteristics, corrosion resistance, in vitro cytocompatibility, and deformation adaptability of the composite coating to the stent were evaluated. The MAO-PBAT composite coating demonstrated a low corrosion current density (Icorr = 2.381 ×10-8 A/cm2), three orders of magnitude lower than that of unmodified EK30 magnesium alloy. Live/dead cell staining results confirmed that the composite coating exhibited good cytocompatibility with human aortic endothelial cells (HAECs) and human aortic smooth muscle cells (HASMCs). Observations of the stent treated with the composite coating during crimping and expansion showed that the composite coating possessed excellent deformation adaptability. These results indicate that the MAO-PBAT composite coating has significant potential for vascular stent application.
Collapse
Affiliation(s)
- Weisheng Cao
- School of Materials Science and Engineering, Dalian University of Technology, Dalian 116024, China
| | - Siyu Zhu
- School of Stomatology, Dalian Medical University, No. 9 West Section, Lvshun South Road, Dalian 116044, China
| | - Weiqiang Wang
- School of Materials Science and Engineering, Dalian University of Technology, Dalian 116024, China.
| | - Yahui Wang
- School of Materials Science and Engineering, Dalian University of Technology, Dalian 116024, China
| | - Shuaikang Yang
- School of Materials Science and Engineering, Dalian University of Technology, Dalian 116024, China
| | - Haiyang Sun
- Central Hospital of Dalian University of Technology (Dalian Central Hospital), Dalian 116033, China.
| |
Collapse
|
3
|
Han H, Ying X, Chen Q, Fang J, Xu D, Lyu X, Zheng J, Zou L, Luo Q, Hu N. Monitoring of inflammatory preterm responses via myometrial cell based multimodal electrophysiological and optical biosensing platform. Biosens Bioelectron 2025; 274:117197. [PMID: 39874921 DOI: 10.1016/j.bios.2025.117197] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Revised: 01/16/2025] [Accepted: 01/23/2025] [Indexed: 01/30/2025]
Abstract
Preterm birth (PTB) remains a leading cause of neonatal morbidity and mortality, with inflammation-induced PTB posing a significant challenge due to its complex pathophysiology. To address this, we developed an in vitro platform utilizing hTERT-immortalized human myometrial (hTERT-HM) cells integrated with a multielectrode array (MEA) biosensing system and optical calcium imaging. Compared to primary uterine myometrial cells, hTERT-HM cells exhibit superior reproducibility, high scalability, and convenient manipulation, facilitating the consistent and large-scale investigations. This advanced system facilitates simultaneous real-time monitoring of electrophysiological activity and intracellular calcium transient, providing detailed insights into uterine cell behavior during inflammatory PTB. Our study revealed that oxytocin (OT) induces regular contractions in hTERT-HM cells, and the synergistic effect of OT and lipopolysaccharide (LPS) disrupts electrophysiological patterns and calcium signaling, closely mimicking the pathophysiology of inflammation-induced PTB. Meanwhile, magnesium sulfate is validated to effectively suppress OT-induced calcium release and mitigate LPS-triggered irregular electrophysiological signals. By integrating advanced biosensing technologies and advantages of hTERT-HM cells, this platform offers a reliable, reproducible model to investigate the mechanisms of inflammation-driven PTB and further develop targeted therapeutic interventions.
Collapse
Affiliation(s)
- Haote Han
- Key Laboratory of Reproductive Genetics (Ministry of Education) and Department of Obstetrics, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, 310006, China; Department of Chemistry, ZJU-Hangzhou Global Scientific and Technological Innovation Center, School of Medicine, Zhejiang University, Hangzhou, 310058, China
| | - Xia Ying
- Key Laboratory of Reproductive Genetics (Ministry of Education) and Department of Obstetrics, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, 310006, China
| | - Qiaoqiao Chen
- Key Laboratory of Reproductive Genetics (Ministry of Education) and Department of Obstetrics, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, 310006, China
| | - Jiaru Fang
- Department of Chemistry, ZJU-Hangzhou Global Scientific and Technological Innovation Center, School of Medicine, Zhejiang University, Hangzhou, 310058, China; Department of Neurology, The Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, 322000, China
| | - Dongxin Xu
- Department of Chemistry, ZJU-Hangzhou Global Scientific and Technological Innovation Center, School of Medicine, Zhejiang University, Hangzhou, 310058, China; General Surgery Department, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Children's Health, Hangzhou, 310052, China
| | - Xuelian Lyu
- Department of Chemistry, ZJU-Hangzhou Global Scientific and Technological Innovation Center, School of Medicine, Zhejiang University, Hangzhou, 310058, China
| | - Jilin Zheng
- Department of Chemistry, ZJU-Hangzhou Global Scientific and Technological Innovation Center, School of Medicine, Zhejiang University, Hangzhou, 310058, China
| | - Ling Zou
- Department of Chemistry, ZJU-Hangzhou Global Scientific and Technological Innovation Center, School of Medicine, Zhejiang University, Hangzhou, 310058, China.
| | - Qiong Luo
- Key Laboratory of Reproductive Genetics (Ministry of Education) and Department of Obstetrics, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, 310006, China.
| | - Ning Hu
- Department of Chemistry, ZJU-Hangzhou Global Scientific and Technological Innovation Center, School of Medicine, Zhejiang University, Hangzhou, 310058, China; General Surgery Department, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Children's Health, Hangzhou, 310052, China.
| |
Collapse
|
4
|
Escudero C, Vatish M. Review: The potential role of placental extracellular vesicles in blood-brain barrier disruption and neuroinflammation in preeclampsia. Placenta 2025:S0143-4004(25)00104-3. [PMID: 40229181 DOI: 10.1016/j.placenta.2025.04.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/26/2024] [Revised: 03/25/2025] [Accepted: 04/01/2025] [Indexed: 04/16/2025]
Abstract
Preeclampsia is a complex pregnancy disorder characterized by hypertension and multisystem organ damage, notably affecting the liver, kidneys, and brain. Eclampsia, a severe form of preeclampsia, is marked by the sudden onset of generalized tonic-clonic seizures. Brain complications, including eclampsia, are responsible for 60-70 % of preeclampsia-related maternal deaths, particularly in low-income regions. Despite the significant impact of brain complications in preeclampsia, their underlying pathophysiology remains unclear. Evidence suggests that brain edema in preeclampsia and eclampsia results from disruption of the blood-brain barrier (BBB). Although direct analysis of the BBB is challenging, in vitro studies indicate that plasma from women with preeclampsia can compromise the BBB, with the specific circulating factors involved still unidentified. Among the potential culprits, recent findings highlight placental-derived small extracellular vesicles (PDsEVs) as key players in BBB disruption observed in preeclampsia. This review examines the role of PDsEVs in the pathophysiology of brain edema associated with preeclampsia, emphasizing areas for future research, including neuroinflammation and neuron dysfunction. Additionally, we discuss the protective role of magnesium sulfate in these processes.
Collapse
Affiliation(s)
- Carlos Escudero
- Vascular Physiology Laboratory, Department of Basic Sciences, Universidad del Bío-Bío, Chillán, Chile; Nuffield Department of Women's & Reproductive Health. University of Oxford, Women's Centre, John Radcliffe Hospital, Oxford, OX3 9DU, United Kingdom; Group of Research and Innovation in Vascular Health (GRIVAS Health), Chillan, Chile.
| | - Manu Vatish
- Nuffield Department of Women's & Reproductive Health. University of Oxford, Women's Centre, John Radcliffe Hospital, Oxford, OX3 9DU, United Kingdom
| |
Collapse
|
5
|
Laragione T, Harris C, Gulko PS. Magnesium Supplementation Modifies Arthritis Synovial and Splenic Transcriptomic Signatures Including Ferroptosis and Cell Senescence Biological Pathways. Nutrients 2024; 16:4247. [PMID: 39683640 DOI: 10.3390/nu16234247] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2024] [Revised: 12/02/2024] [Accepted: 12/05/2024] [Indexed: 12/18/2024] Open
Abstract
BACKGROUND Rheumatoid arthritis (RA) is a common systemic autoimmune inflammatory disease that can cause joint damage. We have recently reported that oral magnesium supplementation significantly reduces disease severity and joint damage in models of RA. METHODS In the present study, we analyzed the transcriptome of spleens and synovial tissues obtained from mice with KRN serum-induced arthritis (KSIA) consuming either a high Mg supplemented diet (Mg2800; n = 7) or a normal diet (Mg500; n = 7). Tissues were collected at the end of a 15-day KSIA experiment. RNA was extracted and used for sequencing and analyses. RESULTS There was an enrichment of differentially expressed genes (DEGs) belonging to Reactome and Gene Ontology (GO) pathways implicated in RA pathogenesis such as RHO GTPases, the RUNX1 pathway, oxidative stress-induced senescence, and the senescence-associated secretory phenotype. Actc1 and Nr4a3 were among the genes with the highest expression, while Krt79 and Ffar2 were among the genes with the lowest expression in synovial tissues of the Mg2800 group compared with the Mg500 group. Spleens had an enrichment for the metabolism of folate and pterines and the HSP90 chaperone cycle for the steroid hormone receptor. CONCLUSIONS We describe the tissue transcriptomic consequences of arthritis-protecting Mg supplementation in KSIA mice. These results show that oral Mg supplementation may interfere with the response to oxidative stress and senescence and other processes known to participate in RA pathogenesis. We provide new evidence supporting the disease-suppressing effect of increased Mg intake in arthritis and its potential to become a new addition to the therapeutic options for RA and other autoimmune and inflammatory diseases.
Collapse
Affiliation(s)
- Teresina Laragione
- Division of Rheumatology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Carolyn Harris
- Division of Rheumatology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Pércio S Gulko
- Division of Rheumatology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| |
Collapse
|
6
|
Zeng T, Lei GL, Yu ML, Zhang TY, Wang ZB, Wang SZ. The role and mechanism of various trace elements in atherosclerosis. Int Immunopharmacol 2024; 142:113188. [PMID: 39326296 DOI: 10.1016/j.intimp.2024.113188] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Revised: 09/13/2024] [Accepted: 09/13/2024] [Indexed: 09/28/2024]
Abstract
Atherosclerosis is a slow and complex disease that involves various factors, including lipid metabolism disorders, oxygen-free radical production, inflammatory cell infiltration, platelet adhesion and aggregation, and local thrombosis. Trace elements play a crucial role in human health. Many trace elements, especially metallic ones, not only maintain the normal functions of organs but also participate in basic metabolic processes. The latest studies have revealed a close correlation between trace elements and the occurrence and progression of atherosclerosis. The imbalance of these trace elements can induce atherosclerosis or accelerate its progression through various mechanisms, which poses a significant threat to human health. Therefore, exploring the specific mechanism of trace elements on atherosclerosis is highly significant. In this review, we summarized the roles and mechanisms of iron, copper, zinc, magnesium, and selenium homeostasis and imbalance in atherosclerosis development, in order to identify novel targets and therapeutic strategies for treating atherosclerosis.
Collapse
Affiliation(s)
- Tao Zeng
- Institute of Pharmacy and Pharmacology, School of Pharmaceutical Sciences, Hengyang Medical School, University of South China, Hengyang 421001, China; Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang 421001, China
| | - Guan-Lan Lei
- Institute of Pharmacy and Pharmacology, School of Pharmaceutical Sciences, Hengyang Medical School, University of South China, Hengyang 421001, China; Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang 421001, China
| | - Mei-Ling Yu
- Institute of Pharmacy and Pharmacology, School of Pharmaceutical Sciences, Hengyang Medical School, University of South China, Hengyang 421001, China; Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang 421001, China
| | - Ting-Yu Zhang
- Institute of Pharmacy and Pharmacology, School of Pharmaceutical Sciences, Hengyang Medical School, University of South China, Hengyang 421001, China; Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang 421001, China
| | - Zong-Bao Wang
- Institute of Pharmacy and Pharmacology, School of Pharmaceutical Sciences, Hengyang Medical School, University of South China, Hengyang 421001, China; Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang 421001, China.
| | - Shu-Zhi Wang
- Institute of Pharmacy and Pharmacology, School of Pharmaceutical Sciences, Hengyang Medical School, University of South China, Hengyang 421001, China; Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang 421001, China.
| |
Collapse
|
7
|
Li S, Chen Z, Yu H, Chang W, Zhou J, Wu G, Sun X, Sun H, Wang K. Association of magnesium deficiency scores with risk of rheumatoid arthritis and osteoarthritis in adults: a cross-sectional population-based study. Clin Rheumatol 2024; 43:3973-3982. [PMID: 39453544 DOI: 10.1007/s10067-024-07203-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Revised: 10/07/2024] [Accepted: 10/17/2024] [Indexed: 10/26/2024]
Abstract
BACKGROUND The magnesium depletion score (MDS) is a scoring system developed to predict magnesium deficiency based on pathophysiological factors that affect renal reabsorption. The relationship between systemic magnesium status and arthritis is unclear. The purpose of this study was to determine the association between the MDS and rheumatoid arthritis (RA) as well as osteoarthritis (OA). METHODS This study was conducted through a cross-sectional survey of 20,513 adults aged ≥ 20 years who participated in NHANES from 2007 to 2018. The four dimensions of the MDS included diuretics, proton pump inhibitors, glomerular filtration rate, and excessive alcohol consumption. Univariate and multivariable-weighted logistic regression were used to assess the associations between MDS and RA/OA, and a test for trend was performed to analyze the presence of a dose-response relationship. Subgroup analyses and interaction tests were performed according to confounders. RESULTS After adjustment for all covariates, we found a graded dose-response relationship between MDS and RA or OA. When MDS was considered as a continuous variable, each onefold increase in MDS was associated with a 1.21-fold increase in the odds of having RA (OR = 1.21, 1.10, 1.33) and a 1.12-fold increase in the odds of having OA (OR = 1.12, 1.04, 1.21). There was an interaction of sex in the effect of MDS on RA (Pinteraction = 0.004) and age in the effect of MDS on OA (Pinteraction = 0.006). In addition, these associations were further confirmed in sensitivity and subgroup analyses. CONCLUSIONS Our study identified significant dose-response associations between MDS and both RA and OA. More biological mechanisms are needed in the future to validate and clarify the results of this study.
Collapse
Affiliation(s)
- Shuxiang Li
- Articular Orthopaedics, The Third Affiliated Hospital of Soochow University, Changzhou, 213003, Jiangsu, China
| | - Zhuo Chen
- Articular Orthopaedics, The Third Affiliated Hospital of Soochow University, Changzhou, 213003, Jiangsu, China
| | - Haoyun Yu
- Suzhou Medical College, Soochow University, Suzhou, 215000, Jiangsu, China
| | - Wenliao Chang
- Articular Orthopaedics, The Third Affiliated Hospital of Soochow University, Changzhou, 213003, Jiangsu, China
| | - Jian Zhou
- Articular Orthopaedics, The Third Affiliated Hospital of Soochow University, Changzhou, 213003, Jiangsu, China
| | - Guofeng Wu
- Articular Orthopaedics, The Third Affiliated Hospital of Soochow University, Changzhou, 213003, Jiangsu, China
| | - Xiaoliang Sun
- Articular Orthopaedics, The Third Affiliated Hospital of Soochow University, Changzhou, 213003, Jiangsu, China
| | - Han Sun
- Articular Orthopaedics, The Third Affiliated Hospital of Soochow University, Changzhou, 213003, Jiangsu, China.
| | - Kun Wang
- Articular Orthopaedics, The Third Affiliated Hospital of Soochow University, Changzhou, 213003, Jiangsu, China.
| |
Collapse
|
8
|
Esmaeily Z, Abaj F, Naeini Z, Alvandi E, Rafiee M, Koohdani F. Dietary acid load adopts the effect of ApoB ins/del genetic variant (rs11279109) on obesity trait, cardiovascular markers, lipid profile, and serum leptin level among patients with diabetes: a cross-sectional study. Sci Rep 2024; 14:25650. [PMID: 39465244 PMCID: PMC11514203 DOI: 10.1038/s41598-024-75352-x] [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: 05/05/2023] [Accepted: 10/04/2024] [Indexed: 10/29/2024] Open
Abstract
ApoB insertion/deletion (ins/del) genetic variant (rs11279109) is thought to be related to cardio-metabolic markers and obesity. This association has the potential to be modified by dietary patterns. Since the majority of studies concerned the role of dietary acid load (DAL) or ApoB in type 2 diabetes mellitus (T2DM) and its complications independently, and due to the insufficient data regarding the possible interactions between ApoB genetic variants and DAL on anthropometric and metabolic markers, we aimed to study the interaction between this genetic variant and dietary acid load (DAL) on cardio-metabolic markers, along with leptin among Iranian individuals with T2DM. 700 T2DM patients were randomly recruited. A validated semi-quantitative food frequency questionnaire was used for DAL calculation including potential renal acid load (PRAL) and net-endogenous acid production (NEAP). The polymerase chain reaction was used for genotyping the ApoB ins/del (rs11279109). The general linear model was applied to find the interactions in the crude and adjusted models. Patients with del/del genotype (rs11279109) with high PRAL intake have lower low-density lipoprotein cholesterol (LDL-C) (Pinteraction = 0.004), LDL/HDL ratio (Pinteraction = 0.02), total cholesterol (TC) (Pinteraction = 0.04), triglyceride (TG) (Pinteraction = 0.04), leptin (Pinteraction = 0.04) and interleukin-18 (IL-18) (Pinteraction = 0.04). Moreover, the interaction of gene and DAL in the PRAL method on TG concentration (P = 0.04), waist circumference (WC) (P = 0.04), and LDL/HDL ratio (P = 0.04) were significant. Eventually, a positive relationship was observed between the presence of the del/del genotype (rs11279109) and higher levels of TG, TC, LDL-C, IL-18, and LDL/HDL, in individuals with lower adherence to DAL, after adjusting for various covariates. Further studies are needed to investigate and confirm these findings.
Collapse
Affiliation(s)
- Zahra Esmaeily
- Department of Nutrition, School of Public Health, Iran University of Medical Sciences, Tehran, Iran
| | - Faezeh Abaj
- Department of nutrition, Dietetics and Food, School of Clinical Sciences at Monash Health, Monash University, Clayton, Australia
| | - Zeinab Naeini
- Department of Cellular and Molecular Nutrition, School of Nutritional Sciences and Dietetics, Tehran University of Medical Sciences, PO Box: 141556117, Tehran, Iran
| | - Ehsan Alvandi
- School of Medicine, Western Sydney University, Campbelltown, NSW, Australia
| | - Masoumeh Rafiee
- Department of Community Nutrition, School of Nutrition and Food Science, Isfahan University of Medical Sciences (IUMS), Isfahan, Iran.
| | - Fariba Koohdani
- Department of Cellular and Molecular Nutrition, School of Nutritional Sciences and Dietetics, Tehran University of Medical Sciences, PO Box: 141556117, Tehran, Iran.
| |
Collapse
|
9
|
Verlato A, Laragione T, Bin S, Kim RH, Salem F, Gulko PS, Cravedi P. Revised version with tracked changes oral Magnesium reduces levels of pathogenic autoantibodies and skin disease in murine lupus. BMC Immunol 2024; 25:58. [PMID: 39242985 PMCID: PMC11378425 DOI: 10.1186/s12865-024-00650-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2024] [Accepted: 08/27/2024] [Indexed: 09/09/2024] Open
Abstract
BACKGROUND Systemic Lupus Erythematosus (SLE) has a strong genetic susceptibility, but little is known about the impact of diet on disease severity. The Western diet is typically deficient in magnesium (Mg), and given the immunomodulatory effects of Mg, we hypothesized that the low Mg intake increases disease risk and that increasing Mg intake would reduce severity of murine lupus. Here, we placed 12-week old MRL/lpr female lupus mice on a normal (Mg500) or a high (Mg2800) Mg diet for 9 weeks. Urine and blood were collected during the study for quantification of urinary albumin, BUN, anti-dsDNA antibodies, and immune phenotyping. RESULTS MRL/lpr lupus mice on high Mg2800 diet had significantly fewer skin lesions and less severe skin histology score, and reduced levels of pathogenic anti-dsDNA antibodies, compared with the Mg500 group (143.8±75.0 vs. 47.4±36.2 × 106U/ml; P < 0.05). The high Mg2800 group had a nearly two-fold increase in the percentage of CD4+FOXP3+ Treg cells compared to controls (19.9±5.4 vs. 11.4±5.5%; P < 0.05). Treg percentages inversely correlated with the concentration of anti-dsDNA. None of the mice developed arthritis during the observation period and there were no significant differences in weight, proteinuria, BUN or kidney histology. CONCLUSION In conclusion, oral supplementation of Mg has a protective effect in a murine lupus model and may represent an inexpensive and safe adjuvant in the treatment of SLsE.
Collapse
Affiliation(s)
- Alberto Verlato
- Precision Immunology Institute, Translational Transplant Research Center, Icahn School of Medicine at Mount Sinai, One Gustave L Levy Place, Box 1243, New York, NY, 10029, USA
- Renal Unit, Department of Medicine, University Hospital of Verona, Verona, Italy
| | - Teresina Laragione
- Division of Rheumatology, Department of Medicine, Icahn School of Medicine at Mount Sinai, One Gustave L Levy Place, Box 1243, New York, NY, 10029, USA
| | - Sofia Bin
- Precision Immunology Institute, Translational Transplant Research Center, Icahn School of Medicine at Mount Sinai, One Gustave L Levy Place, Box 1243, New York, NY, 10029, USA
- Nephrology, Dialysis and Renal Transplant Unit, IRCCS - Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
- CIRI Scienze della Vita e Tecnologie per la Salute - Alma Mater Studiorum Università di Bologna, Bologna, Italy
| | - Randie H Kim
- Dermatopathology Section, The Kimberley and Eric J. Waldman Department of Dermatology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Fadi Salem
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Jacksonville, FL, USA
| | - Percio S Gulko
- Division of Rheumatology, Department of Medicine, Icahn School of Medicine at Mount Sinai, One Gustave L Levy Place, Box 1243, New York, NY, 10029, USA.
| | - Paolo Cravedi
- Precision Immunology Institute, Translational Transplant Research Center, Icahn School of Medicine at Mount Sinai, One Gustave L Levy Place, Box 1243, New York, NY, 10029, USA.
| |
Collapse
|
10
|
Zaslow SJ, Oliveira-Paula GH, Chen W. Magnesium and Vascular Calcification in Chronic Kidney Disease: Current Insights. Int J Mol Sci 2024; 25:1155. [PMID: 38256228 PMCID: PMC10816532 DOI: 10.3390/ijms25021155] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Revised: 01/12/2024] [Accepted: 01/14/2024] [Indexed: 01/24/2024] Open
Abstract
Magnesium (Mg) plays crucial roles in multiple essential biological processes. As the kidneys are the primary organ responsible for maintaining the blood concentration of Mg, people with chronic kidney disease (CKD) may develop disturbances in Mg. While both hyper- and hypomagnesemia may lead to adverse effects, the consequences associated with hypomagnesemia are often more severe and lasting. Importantly, observational studies have shown that CKD patients with hypomagnesemia have greater vascular calcification. Vascular calcification is accelerated and contributes to a high mortality rate in the CKD population. Both in vitro and animal studies have demonstrated that Mg protects against vascular calcification via several potential mechanisms, such as inhibiting the formation of both hydroxyapatite and pathogenic calciprotein particles as well as limiting osteogenic differentiation, a process in which vascular smooth muscle cells in the media layer of the arteries transform into bone-like cells. These preclinical findings have led to several important clinical trials that have investigated the effects of Mg supplementation on vascular calcification in people with CKD. Interestingly, two major clinical studies produced contradictory findings, resulting in a state of equipoise. This narrative review provides an overview of our current knowledge in the renal handling of Mg in health and CKD and the underlying mechanisms by which Mg may protect against vascular calcification. Lastly, we evaluate the strength of evidence from clinical studies on the efficacy of Mg supplementation and discuss future research directions.
Collapse
Affiliation(s)
- Shari J. Zaslow
- Department of Medicine, Nephrology Division, Albert Einstein College of Medicine, Bronx, NY 10461, USA
- The Robert Larner, M.D. College of Medicine, University of Vermont, Burlington, VT 05405, USA
| | - Gustavo H. Oliveira-Paula
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
- Wilf Family Cardiovascular Research Institute, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Wei Chen
- Department of Medicine, Nephrology Division, Albert Einstein College of Medicine, Bronx, NY 10461, USA
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| |
Collapse
|
11
|
Batista PR, de Alencar Silva A, Mikevely de Sena Bastos C, de Souza Borges A, Dias FJ, Ramon Dos Santos Pereira L, Tavares de Sousa Machado S, de Araújo Delmondes G, Kerntopf MR, Alencar de Menezes IR, Barbosa R. Vasorelaxant effect of (E,E)-farnesol in human umbilical vein ex vivo assays. Chem Biol Interact 2023; 386:110746. [PMID: 37816450 DOI: 10.1016/j.cbi.2023.110746] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2023] [Revised: 09/01/2023] [Accepted: 09/22/2023] [Indexed: 10/12/2023]
Abstract
(E,E)-farnesol is a sesquiterpene acyclic alcohol produced by bacteria, protozoa, fungi, plants, and animals. The literature describes its applications in food, pharmaceutical, and cosmetic industries, and also in the pharmacological context with a vasorelaxant effect. However, its effects on human umbilical vessels remain poorly investigated. Thus, this study aims to investigate, in a new way, the vasorelaxant effect of (E,E)-farnesol in human umbilical veins (HUV) from healthy donors. Rings obtained from isolated HUV were suspended in an organ bath to record their isometric tension in different experimental sections. (E,E)-farnesol (1 μmol/L to 1 mmol/L) promoted vasorelaxant effect in venous preparations contracted by depolarization (KCl 60 mmol/L) or pharmacological agonism (5-HT 10 μmol/L), with EC50 values of 239.9 μmol/L and 424 μmol/L, respectively. In calcium-free solution, this effect was also observable. (E,E)-farnesol was able to suppress contractions evoked by CaCl2 and BaCl2 suggesting a blockade of voltage-dependent (especially L-type) calcium channels. The vasorelaxant efficacy and potency of (E,E)-farnesol were affected in the presence of tetraethylammonium (1 and 10 mmol/L), glibenclamide (10 μmol/L) and BaCl2 (1 mmol/L) indicating a possible involvement of potassium channels (BKCa, KATP and KIR) in this effect. Our data suggest that (E,E)-farnesol has a promising potential to be applicable as a vasodilator in hypertensive conditions in pregnancy that alter HUV reactivity.
Collapse
Affiliation(s)
- Paulo Ricardo Batista
- Biological Chemistry Department, Pimenta Campus, Regional University of Cariri, Crato, 63105-000, Ceará, Brazil; Biological Sciences Department, Physiopharmacology of Excitable Cells Laboratory, Pimenta Campus, Regional University of Cariri, Crato, 63105-000, Ceará, Brazil.
| | - Andressa de Alencar Silva
- Biological Sciences Department, Physiopharmacology of Excitable Cells Laboratory, Pimenta Campus, Regional University of Cariri, Crato, 63105-000, Ceará, Brazil; Higher Institute of Biomedical Sciences, State University of Ceará, Fortaleza, 60714-903, Ceará, Brazil.
| | - Carla Mikevely de Sena Bastos
- Biological Chemistry Department, Pimenta Campus, Regional University of Cariri, Crato, 63105-000, Ceará, Brazil; Biological Sciences Department, Physiopharmacology of Excitable Cells Laboratory, Pimenta Campus, Regional University of Cariri, Crato, 63105-000, Ceará, Brazil.
| | - Alex de Souza Borges
- Biological Chemistry Department, Pimenta Campus, Regional University of Cariri, Crato, 63105-000, Ceará, Brazil; Biological Sciences Department, Physiopharmacology of Excitable Cells Laboratory, Pimenta Campus, Regional University of Cariri, Crato, 63105-000, Ceará, Brazil.
| | - Francisco Junio Dias
- Biological Sciences Department, Physiopharmacology of Excitable Cells Laboratory, Pimenta Campus, Regional University of Cariri, Crato, 63105-000, Ceará, Brazil.
| | - Luiz Ramon Dos Santos Pereira
- Biological Sciences Department, Physiopharmacology of Excitable Cells Laboratory, Pimenta Campus, Regional University of Cariri, Crato, 63105-000, Ceará, Brazil.
| | - Sara Tavares de Sousa Machado
- Biological Sciences Department, Physiopharmacology of Excitable Cells Laboratory, Pimenta Campus, Regional University of Cariri, Crato, 63105-000, Ceará, Brazil.
| | | | - Marta Regina Kerntopf
- Biological Chemistry Department, Pimenta Campus, Regional University of Cariri, Crato, 63105-000, Ceará, Brazil.
| | | | - Roseli Barbosa
- Biological Chemistry Department, Pimenta Campus, Regional University of Cariri, Crato, 63105-000, Ceará, Brazil; Biological Sciences Department, Physiopharmacology of Excitable Cells Laboratory, Pimenta Campus, Regional University of Cariri, Crato, 63105-000, Ceará, Brazil.
| |
Collapse
|
12
|
Laragione T, Harris C, Azizgolshani N, Beeton C, Bongers G, Gulko PS. Magnesium increases numbers of Foxp3+ Treg cells and reduces arthritis severity and joint damage in an IL-10-dependent manner mediated by the intestinal microbiome. EBioMedicine 2023; 92:104603. [PMID: 37201335 PMCID: PMC10203746 DOI: 10.1016/j.ebiom.2023.104603] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Revised: 03/10/2023] [Accepted: 04/19/2023] [Indexed: 05/20/2023] Open
Abstract
BACKGROUND Rheumatoid arthritis (RA) is a common autoimmune disease with emerging environmental and microbiome risk factors. The western diet is typically deficient in magnesium (Mg), and there is some evidence suggesting that Mg may have anti-inflammatory properties. But the actual role of Mg supplementation in arthritis or in T cell subsets has not been explored. METHODS We investigated the role of a high Mg diet in two different mouse models of RA induced with the KRN serum, and collagen-induced arthritis. We also characterized the phenotypes of splenocytes, gene expression, and an extensive intestinal microbiome analyses including fecal material transplantation (FMT). FINDINGS The high Mg diet group was significantly protected with reduced arthritis severity and joint damage, and reduced expression of IL-1β, IL-6, and TNFα. The high Mg group also had increased numbers of Foxp3+ Treg cells and IL-10-producing T cells. The high Mg protective effect disappeared in IL-10 knockout mice. FMT from the high Mg diet mice recreated the phenotypes seen in the diet-treated mice, with reduced arthritis severity, increased Foxp3+ Treg, and increased IL-10-producing T cells. Intestinal microbiome analyses using 16S rDNA sequencing revealed diet-specific changes, including reduced levels of RA-associated Prevotella in the high Mg group, while increasing levels of Bacteroides and other bacteria associated with increased production of short-chain fatty acids. Metagenomic analyses implicated additional pathways including L-tryptophan biosynthesis and arginine deiminase. INTERPRETATION We describe a new role for Mg in suppressing arthritis, in expanding Foxp3+ T reg cells and in the production of IL-10, and show that these effects are mediated by the intestinal microbiome. Our discoveries suggest a novel strategy for modifying the intestinal microbiome to treat RA and other autoimmune and inflammatory diseases. FUNDING None.
Collapse
Affiliation(s)
- Teresina Laragione
- Division of Rheumatology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, United States
| | - Carolyn Harris
- Division of Rheumatology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, United States
| | - Nasim Azizgolshani
- Division of Rheumatology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, United States
| | - Christine Beeton
- Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, TX, 77030, United States
| | - Gerold Bongers
- Microbiome Translational Center, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, United States
| | - Percio S Gulko
- Division of Rheumatology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, United States.
| |
Collapse
|
13
|
Severino P, D’Amato A, Prosperi S, Myftari V, Labbro Francia A, Önkaya M, Notari C, Papisca I, Canuti ES, Yarden Revivo M, Birtolo LI, Celli P, Galardo G, Maestrini V, d’Ettorre G, Mancone M, Fedele F. The Mutual Relationship among Cardiovascular Diseases and COVID-19: Focus on Micronutrients Imbalance. Nutrients 2022; 14:3439. [PMID: 36014944 PMCID: PMC9416353 DOI: 10.3390/nu14163439] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2022] [Revised: 08/12/2022] [Accepted: 08/15/2022] [Indexed: 12/16/2022] Open
Abstract
Micronutrients are ions and vitamins humbly required by the human body. They play a main role in several physiological mechanisms and their imbalance is strongly associated with potentially-fatal complications. Micronutrient imbalance is associated with many cardiovascular diseases, such as arrythmias, heart failure, and ischemic heart disease. It has been also observed in coronavirus disease 2019 (COVID-19), particularly in most severe patients. The relationship between cardiovascular diseases and COVID-19 is mutual: the latter triggers cardiovascular disease onset and worsening while patients with previous cardiovascular disease may develop a more severe form of COVID-19. In addition to the well-known pathophysiological mechanisms binding COVID-19 and cardiovascular diseases together, increasing importance is being given to the impact of micronutrient alterations, often present during COVID-19 and able to affect the balance responsible for a good functioning of the cardiovascular system. In particular, hypokalemia, hypomagnesemia, hyponatremia, and hypocalcemia are strongly associated with worse outcome, while vitamin A and D deficiency are associated with thromboembolic events in COVID-19. Thus, considering how frequent the cardiovascular involvement is in patients with COVID-19, and how it majorly affects their prognosis, this manuscript provides a comprehensive review on the role of micronutrient imbalance in the interconnection between COVID-19 and cardiovascular diseases.
Collapse
Affiliation(s)
- Paolo Severino
- Department of Clinical, Internal, Anesthesiology and Cardiovascular Sciences, Sapienza University of Rome, Viale del Policlinico 155, 00161 Rome, Italy
| | - Andrea D’Amato
- Department of Clinical, Internal, Anesthesiology and Cardiovascular Sciences, Sapienza University of Rome, Viale del Policlinico 155, 00161 Rome, Italy
| | - Silvia Prosperi
- Department of Clinical, Internal, Anesthesiology and Cardiovascular Sciences, Sapienza University of Rome, Viale del Policlinico 155, 00161 Rome, Italy
| | - Vincenzo Myftari
- Department of Clinical, Internal, Anesthesiology and Cardiovascular Sciences, Sapienza University of Rome, Viale del Policlinico 155, 00161 Rome, Italy
| | - Aurora Labbro Francia
- Department of Clinical, Internal, Anesthesiology and Cardiovascular Sciences, Sapienza University of Rome, Viale del Policlinico 155, 00161 Rome, Italy
| | - Merve Önkaya
- Department of Clinical, Internal, Anesthesiology and Cardiovascular Sciences, Sapienza University of Rome, Viale del Policlinico 155, 00161 Rome, Italy
| | - Claudia Notari
- Department of Clinical, Internal, Anesthesiology and Cardiovascular Sciences, Sapienza University of Rome, Viale del Policlinico 155, 00161 Rome, Italy
| | - Ilaria Papisca
- Department of Clinical, Internal, Anesthesiology and Cardiovascular Sciences, Sapienza University of Rome, Viale del Policlinico 155, 00161 Rome, Italy
| | - Elena Sofia Canuti
- Department of Clinical, Internal, Anesthesiology and Cardiovascular Sciences, Sapienza University of Rome, Viale del Policlinico 155, 00161 Rome, Italy
| | - Mia Yarden Revivo
- Department of Clinical, Internal, Anesthesiology and Cardiovascular Sciences, Sapienza University of Rome, Viale del Policlinico 155, 00161 Rome, Italy
| | - Lucia Ilaria Birtolo
- Department of Clinical, Internal, Anesthesiology and Cardiovascular Sciences, Sapienza University of Rome, Viale del Policlinico 155, 00161 Rome, Italy
| | - Paola Celli
- Anesthesiology and Intensive Care Unit, Sapienza University of Rome, Policlinico Umberto I, 00161 Rome, Italy
| | - Gioacchino Galardo
- Medical Emergency Unit, Sapienza University of Rome, Policlinico Umberto I, 00185 Rome, Italy
| | - Viviana Maestrini
- Department of Clinical, Internal, Anesthesiology and Cardiovascular Sciences, Sapienza University of Rome, Viale del Policlinico 155, 00161 Rome, Italy
| | - Gabriella d’Ettorre
- Department of Public Health and Infectious Diseases, Sapienza University of Rome, 00185 Rome, Italy
| | - Massimo Mancone
- Department of Clinical, Internal, Anesthesiology and Cardiovascular Sciences, Sapienza University of Rome, Viale del Policlinico 155, 00161 Rome, Italy
| | - Francesco Fedele
- Department of Clinical, Internal, Anesthesiology and Cardiovascular Sciences, Sapienza University of Rome, Viale del Policlinico 155, 00161 Rome, Italy
| |
Collapse
|
14
|
Liang L, Song D, Wu K, Ouyang Z, Huang Q, Lei G, Zhou K, Xiao J, Wu H. Sequential activation of M1 and M2 phenotypes in macrophages by Mg degradation from Ti-Mg alloy for enhanced osteogenesis. Biomater Res 2022; 26:17. [PMID: 35484564 PMCID: PMC9052665 DOI: 10.1186/s40824-022-00262-w] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Accepted: 03/26/2022] [Indexed: 01/04/2023] Open
Abstract
BACKGROUND Even though the modulatory effects of Magnisum (Mg) and its alloys on bone-healing cells have been widely investigated during the last two decades, relatively limited attention has been paid on their inflammation-modulatory properties. Understanding the activation process of macrophages in response to the dynamic degradation process of Mg as well as the relationship between macrophage phenotypes and their osteogenic potential is critical for the design and development of advanced Mg-based or Mg-incorporated biomaterials. METHODS In this work, a Ti-0.625 Mg (wt.%) alloy fabricated by mechanical alloying (MA) and subsequent spark plasma sintering (SPS) was employed as a material model to explore the inflammatory response and osteogenic performance in vitro and in vivo by taking pure Ti as the control. The data analysis was performed following Student's t-test. RESULTS The results revealed that the macrophages grown on the Ti-0.625 Mg alloy underwent sequential activation of M1 and M2 phenotypes during a culture period of 5 days. The initially increased environmental pH (~ 8.03) was responsible for the activation of M1 macrophages, while accumulated Mg2+ within cells contributed to the lateral M2 phenotype activation. Both M1 and M2 macrophages promoted osteoblast-like SaOS-2 cell maturation. In vivo experiment further showed the better anti-inflammatory response, regenerative potentiality and thinner fibrous tissue layer for the Ti-0.625 Mg alloy than pure Ti. CONCLUSION The results highlighted the roles of Mg degradation in the Ti-0.625 Mg alloy on the sequential activation of macrophage phenotypes and the importance of modulating M1-to-M2 transition in macrophage phenotypes for the design and development of inflammation-modulatory biomaterials.
Collapse
Affiliation(s)
- Luxin Liang
- State Key Laboratory of Powder Metallurgy, Central South University, Changsha, 410083, People's Republic of China.,Department of Orthopedics, The Second Xiangya Hospital, Central South University, Changsha, 410011, People's Republic of China
| | - Deye Song
- Department of Orthopedics, The Second Xiangya Hospital, Central South University, Changsha, 410011, People's Republic of China
| | - Kai Wu
- Department of Rehabilitation, Xiangya Hospital, Central South University, Changsha, 410008, People's Republic of China
| | - Zhengxiao Ouyang
- Department of Orthopedics, The Second Xiangya Hospital, Central South University, Changsha, 410011, People's Republic of China
| | - Qianli Huang
- State Key Laboratory of Powder Metallurgy, Central South University, Changsha, 410083, People's Republic of China. .,Foshan (Southern China) Institute for New Materials, Foshan, 528200, People's Republic of China.
| | - Guanghua Lei
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, 410008, People's Republic of China
| | - Kun Zhou
- School of Mechanical and Aerospace Engineering, Nanyang Technological University, Singapore, 639798, Singapore
| | - Jian Xiao
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha, 410008, People's Republic of China. .,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, People's Republic of China.
| | - Hong Wu
- State Key Laboratory of Powder Metallurgy, Central South University, Changsha, 410083, People's Republic of China.
| |
Collapse
|
15
|
Rajendran P, AlZahrani A. Fucoxanthin suppresses OxLDL-induced inflammation via activation of Nrf2 and inhibition of NF-κB signaling. Asian Pac J Trop Biomed 2022. [DOI: 10.4103/2221-1691.343388] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
|
16
|
León J, Acurio J, Bergman L, López J, Karin Wikström A, Torres-Vergara P, Troncoso F, Castro FO, Vatish M, Escudero C. Disruption of the Blood-Brain Barrier by Extracellular Vesicles From Preeclampsia Plasma and Hypoxic Placentae: Attenuation by Magnesium Sulfate. Hypertension 2021; 78:1423-1433. [PMID: 34601964 DOI: 10.1161/hypertensionaha.121.17744] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
[Figure: see text].
Collapse
Affiliation(s)
- José León
- Vascular Physiology Laboratory, Department of Basic Sciences, Universidad del Bío-Bío, Chillán, Chile (J. Leon, J.A., J. Lopez, F.T., C.E.)
- Escuela de Enfermería, Facultad de Salud, Universidad Santo Tomás, Los Ángeles, Chile (J. Leon,.)
| | - Jesenia Acurio
- Vascular Physiology Laboratory, Department of Basic Sciences, Universidad del Bío-Bío, Chillán, Chile (J. Leon, J.A., J. Lopez, F.T., C.E.)
- Group of Research and Innovation in Vascular Health (Group of Research and Innovation in Vascular Health), Chillan, Chile (J.A., F.T., C.E., P.T.-V.)
| | - Lina Bergman
- Department of Women's and Children's Health, Uppsala University, Sweden (L.B., A.K.W.)
- Department of Clinical Sciences, Sahlgrenska Academy, Gothenburg University, Sweden (L.B.)
- Department of Obstetrics and Gynecology, Stellenbosch University, South Africa (L.B.)
| | - Juán López
- Vascular Physiology Laboratory, Department of Basic Sciences, Universidad del Bío-Bío, Chillán, Chile (J. Leon, J.A., J. Lopez, F.T., C.E.)
| | - Anna Karin Wikström
- Department of Women's and Children's Health, Uppsala University, Sweden (L.B., A.K.W.)
| | - Pablo Torres-Vergara
- Group of Research and Innovation in Vascular Health (Group of Research and Innovation in Vascular Health), Chillan, Chile (J.A., F.T., C.E., P.T.-V.)
- Department of Pharmacy, Faculty of Pharmacy, University of Concepción, Chile (P.T.-V.)
| | - Felipe Troncoso
- Vascular Physiology Laboratory, Department of Basic Sciences, Universidad del Bío-Bío, Chillán, Chile (J. Leon, J.A., J. Lopez, F.T., C.E.)
- Group of Research and Innovation in Vascular Health (Group of Research and Innovation in Vascular Health), Chillan, Chile (J.A., F.T., C.E., P.T.-V.)
| | - Fidel Ovidio Castro
- Department of Animal Science, Faculty of Veterinary Sciences, Universidad de Concepcion, Chillan, Chile (F.O.C.)
| | - Manu Vatish
- Nuffield Department of Women's & Reproductive Health. University of Oxford, Women's Centre, John Radcliffe Hospital, United Kingdom (M.V.)
| | - Carlos Escudero
- Vascular Physiology Laboratory, Department of Basic Sciences, Universidad del Bío-Bío, Chillán, Chile (J. Leon, J.A., J. Lopez, F.T., C.E.)
- Group of Research and Innovation in Vascular Health (Group of Research and Innovation in Vascular Health), Chillan, Chile (J.A., F.T., C.E., P.T.-V.)
| |
Collapse
|
17
|
Zamir A, Ben-Zeev T, Hoffman JR. Manipulation of Dietary Intake on Changes in Circulating Testosterone Concentrations. Nutrients 2021; 13:3375. [PMID: 34684376 PMCID: PMC8538516 DOI: 10.3390/nu13103375] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2021] [Revised: 09/22/2021] [Accepted: 09/23/2021] [Indexed: 12/22/2022] Open
Abstract
Elevations in the circulating concentration of androgens are thought to have a positive effect on the anabolic processes leading to improved athletic performance. Anabolic-androgenic steroids have often been used by competitive athletes to augment this effect. Although there has been concerted effort on examining how manipulating training variables (e.g., intensity and volume of training) can influence the androgen response to exercise, there has been much less effort directed at understanding how changes in both macronutrient and micronutrient intake can impact the androgen response. Thus, the focus of this review is to examine the effect that manipulating energy and nutrient intake has on circulating concentrations of testosterone and what the potential mechanism is governing these changes.
Collapse
Affiliation(s)
| | | | - Jay R. Hoffman
- Department of Physical Therapy, Faculty of Health Sciences, Ariel University, 40700 Ariel, Israel; (A.Z.); (T.B.-Z.)
| |
Collapse
|
18
|
Li W, Jian X, Zou Y, Wu L, Huang H, Li H, Hu D, Yu B. The Fabrication of a Gellan Gum-Based Hydrogel Loaded With Magnesium Ions for the Synergistic Promotion of Skin Wound Healing. Front Bioeng Biotechnol 2021; 9:709679. [PMID: 34589471 PMCID: PMC8473818 DOI: 10.3389/fbioe.2021.709679] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Accepted: 08/09/2021] [Indexed: 01/26/2023] Open
Abstract
To accelerate serious skin burn wound healing in a convenient manner, an interpenetrating network of hydrogel consisting of gellan gum and polyacrylamide was synthesized by chemical crosslinking and Mg2+ ion immersion techniques. The prepared Mg2+@PAM/GG hydrogel was characterized by morphology, water vapor loss, swelling ratio, rheological properties, tensile mechanical, biocompatibility, and flow cytometry study. The results show that Mg2+@PAM/GG hydrogel's mechanical strength could be enhanced by the dual network structure and physical crosslinking agent Mg2+ ions. In addition, the tension strength of Mg2+@PAM/GG hydrogel is obviously increased from 86 to 392 kPa, the elongation at break increased from 84 to 231%, and crosslinking density N increased from 4.3 to 7.2 mol/m3 compared with pure GG hydrogel. The cumulative release curve of Mg2+ ions shows that the multiple release mechanism of Mg2+ ions belong to non-Fick's diffusion. Meanwhile, in vitro experiments show that Mg2+@PAM/GG double network hydrogel has favorable proliferation and an NF-κB pathway inhibition property for fibroblast cells. Finally, the healing effect of the Mg2+@PAM/GG was evaluated in a rat full-thickness burn model. The animal study demonstrates that Mg2+@PAM/GG could accelerate the healing efficiency in case of the sustained-released Mg2+ ions in wound beds. Considering this excellent performance, this convenient prepared hydrogel has great potential as a commercial application for skin full-thickness burn healing materials.
Collapse
Affiliation(s)
- Wenqiang Li
- Department of Dermatology, Skin Research Institute of Peking University Shenzhen Hospital, Peking University Shenzhen Hospital, Shenzhen, China
- Guangdong Provincial Engineering Technology Research Center for Sports Assistive Devices, Guangzhou Sport University, Guangzhou, China
| | - Xingling Jian
- Department of Dermatology, Skin Research Institute of Peking University Shenzhen Hospital, Peking University Shenzhen Hospital, Shenzhen, China
| | - Yanfen Zou
- Department of Dermatology, Skin Research Institute of Peking University Shenzhen Hospital, Peking University Shenzhen Hospital, Shenzhen, China
| | - Lin Wu
- Department of Dermatology, Skin Research Institute of Peking University Shenzhen Hospital, Peking University Shenzhen Hospital, Shenzhen, China
| | - Haiyan Huang
- Department of Dermatology, Skin Research Institute of Peking University Shenzhen Hospital, Peking University Shenzhen Hospital, Shenzhen, China
| | - Hui Li
- Department of Dermatology, Skin Research Institute of Peking University Shenzhen Hospital, Peking University Shenzhen Hospital, Shenzhen, China
| | - Dandan Hu
- Child Healthcare Department, Guangzhou Women’s and Children’s Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Bo Yu
- Department of Dermatology, Skin Research Institute of Peking University Shenzhen Hospital, Peking University Shenzhen Hospital, Shenzhen, China
| |
Collapse
|
19
|
Intravenous magnesium sulfate for prevention of vancomycin plus piperacillin-tazobactam induced acute kidney injury in critically ill patients: An open-label, placebo-controlled, randomized clinical trial. ACTA ACUST UNITED AC 2021; 29:341-351. [PMID: 34463929 DOI: 10.1007/s40199-021-00411-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Accepted: 08/07/2021] [Indexed: 10/20/2022]
Abstract
BACKGROUND Recent studies have shown an increased risk of acute kidney injury (AKI) induced by vancomycin + piperacillin-tazobactam (VPT) combination. In this study, the efficacy of intravenous magnesium sulfate in prevention of VPT induced AKI in critically ill patients admitted to the ICU has been evaluated. METHODS In an open-label, placebo-controlled, randomized clinical trial, 72 adults (≥ 18 years old) who had indications to receive VPT as empiric therapy were assigned to the magnesium or control group in 1:1 ratio. Concomitant with VPT, intravenous infusion of magnesium sulfate was started for patients in the magnesium group. The target serum level of magnesium was defined 3 mg/dl. Patients in the control group received normal saline as placebo. The target serum level of magnesium was defined 1.9 mg/dl in this group. The study's primary outcome was incidence of AKI during and up to 48 h after the treatment course. Escalation and de-escalation of VPT regimen, duration of hospitalization, length of ICU stay and 28-day mortality were secondary outcomes. RESULTS Thirty patients in each group completed the examination. Five patients in the magnesium group and 11 patients in the control group experienced AKI (p = 0.072). De-escalation of VPT regimen was done approximately in 60% of patients. Duration of hospitalization and length of ICU stay were not statistically different between the groups. Finally, 28-day mortality was 23.33% in each group. Although the incidence of AKI was not statistically different between the groups in unadjusted logistic regression model, it became significant after adjusting for confounding factors [unadjusted model (OR = 0.34; 95% CI: 0.10-1.16, p = 0.084), adjusted model: (OR = 0.26; 95% CI: 0.07-0.96, p = 0.04)]. CONCLUSIONS Administration of magnesium sulfate with the target serum levels around 3 mg/dL reduced the incidence of AKI in critically ill patients who were receiving VPT as empric therapy.
Collapse
|
20
|
Magnesium sulfate ameliorates sepsis-induced diaphragm dysfunction in rats via inhibiting HMGB1/TLR4/NF-κB pathway. Neuroreport 2021; 31:902-908. [PMID: 32558672 PMCID: PMC7368847 DOI: 10.1097/wnr.0000000000001478] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Diaphragm dysfunction could be induced by sepsis with subsequent ventilatory pump failure that is associated with local infiltration of inflammatory factors in the diaphragm. It has been shown that the administration of anticonvulsant agent, magnesium sulfate (MgSO4) could decrease systematic inflammatory response. We recently reported that MgSO4 could inhibit macrophages high mobility group box 1 (HMGB1) secretion that confirms its anti-inflammatory properties. Toll-like receptor 4 (TLR4)/nuclear factor-kappa B (NF-κB) signal pathway appears to be involved in the pathology of septic experimental animal’s inflammatory response and involve in the pathogenic mechanisms of sepsis-induced diaphragm dysfunction. Thus, in this study, we are aiming to explore whether MgSO4 could ameliorate sepsis-induced diaphragm dysfunction via TLR4/NF-κB pathway in a rodent model with controlled mechanical ventilation (CMV) and subsequent septic challenge.
Collapse
|
21
|
Harmon MBA, Heijnen NFL, de Bruin S, Sperna Weiland NH, Meijers JCM, de Boer AM, Schultz MJ, Horn J, Juffermans NP. Induced normothermia ameliorates the procoagulant host response in human endotoxaemia. Br J Anaesth 2021; 126:1111-1118. [PMID: 33896590 PMCID: PMC8258978 DOI: 10.1016/j.bja.2021.02.033] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Revised: 02/01/2021] [Accepted: 02/22/2021] [Indexed: 12/02/2022] Open
Abstract
Background Dysregulation of coagulation occurs commonly in sepsis, ranging from mild coagulopathy with decreased platelets to disseminated intravascular coagulation (DIC). We investigated the effect of induced normothermia on coagulation during lipopolysaccharide (LPS)-induced endotoxaemia in healthy volunteers. Methods Twelve volunteers received an infusion of bacterial lipopolysaccharide (Escherichia coli; 2 ng kg−1) and were assigned to either induced normothermia or control. Induced normothermia to maintain core temperature at 37°C consisted of external surface cooling, cold i.v. fluids, and medication to reduce shivering (buspirone, clonidine, and magnesium sulphate). The primary outcome was the DIC score (International Society on Thrombosis and Haemostasis guideline). Prothrombin time (PT), activated partial thromboplastin time (aPTT), D-dimer, plasma von Willebrand factor (vWf), and rotational thromboelastometry (ROTEM) were measured before and 1, 3, 6, and 8 h after LPS infusion. Differences between groups were tested with a mixed effects model. Results In control subjects, lipopolysaccharide caused a fever, transiently decreased platelet levels and lowered activated partial thromboplastin time, while prolonging prothrombin time and increasing D-Dimer and vWf levels. Normothermia prevented the DIC-score exceeding 4, which occurred in 50% of control subjects. Normothermia also reduced the fall in platelet count by 67x109 L−1([95%CI:27-107]; p=0.002), aPTT (mean difference:3s [95%CI:1-5]; p=0.005) and lowered vWf levels by 89% ([95%CI:6-172]; p=0.03), compared to the fever group. ROTEM measurements were unaffected by lipopolysaccharide. Conclusion In human endotoxaemia, induced normothermia decreases markers of endothelial activation and DIC. Maintaining normothermia may reduce coagulopathy in hyperinflammatory states.
Collapse
Affiliation(s)
- Matthew B A Harmon
- Laboratory of Experimental Intensive Care and Anaesthesiology, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, the Netherlands; Department of Intensive Care Medicine, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, the Netherlands.
| | - Nanon F L Heijnen
- Department of Intensive Care Medicine, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, the Netherlands
| | - Sanne de Bruin
- Laboratory of Experimental Intensive Care and Anaesthesiology, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, the Netherlands; Department of Intensive Care Medicine, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, the Netherlands
| | - Niek H Sperna Weiland
- Department of Anaesthesiology, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, the Netherlands
| | - Joost C M Meijers
- Department of Experimental Vascular Medicine, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, the Netherlands; Department of Molecular and Cellular Haemostasis, Sanquin, Amsterdam, the Netherlands
| | - Anita M de Boer
- Department of Intensive Care Medicine, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, the Netherlands
| | - Marcus J Schultz
- Laboratory of Experimental Intensive Care and Anaesthesiology, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, the Netherlands; Department of Intensive Care Medicine, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, the Netherlands; Mahidol Oxford Research Unit, Mahidol University, Bangkok, Thailand; Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Janneke Horn
- Laboratory of Experimental Intensive Care and Anaesthesiology, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, the Netherlands; Department of Intensive Care Medicine, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, the Netherlands
| | - Nicole P Juffermans
- Laboratory of Experimental Intensive Care and Anaesthesiology, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, the Netherlands; Department of Intensive Care Medicine, OLVG Hospital, Amsterdam, the Netherlands
| |
Collapse
|
22
|
Rahmani H, Khalili H. Prevention of vancomycin-induced nephrotoxicity; an update review of clinical and preclinical studies. Infect Disord Drug Targets 2021; 22:e310321192584. [PMID: 33797371 DOI: 10.2174/1871526521666210331164552] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Revised: 12/14/2020] [Accepted: 12/29/2020] [Indexed: 11/22/2022]
Abstract
PURPOSE Clinical and preclinical evidences regarding new strategies for prevention of vancomycin-induced nephrotoxicity are reviewed. METHODS Evidence from 2014 to end of 2019 was included. Finally, twelve animal studies and one clinical trial were evaluated. RESULTS Although incidence of vancomycin-induced nephrotoxicity was not reduced significantly in the clinical trial, antioxidants reduced incidence of vancomycin-induced nephrotoxicity in preclinical studies. CONCLUSIONS Antioxidants including vitamin C, vitamin E, cilastatin, melatonin, zingerone, rutin, naringenin, saffron, silymarin and dexmedetomidine were nephroprotective against vancomycin-induced nephrotoxicity in preclinical studies. The nephroprotective effects of these antioxidants must be confirmed before routine use in the clinical practice.
Collapse
Affiliation(s)
- Hamid Rahmani
- Department of Clinical Pharmacy, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran. Iran
| | - Hossein Khalili
- Department of Clinical Pharmacy, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran. Iran
| |
Collapse
|
23
|
Xu L, Willumeit-Römer R, Luthringer-Feyerabend BJC. Mesenchymal Stem Cell and Oxygen Modulate the Cocultured Endothelial Cells in the Presence of Magnesium Degradation Products. ACS APPLIED BIO MATERIALS 2021; 4:2398-2407. [DOI: 10.1021/acsabm.0c01289] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Affiliation(s)
- Lei Xu
- Institute of Materials Research, Division for Metallic Biomaterials, Helmholtz-Zentrum Geesthacht (HZG), Geesthacht 21502, Germany
| | - Regine Willumeit-Römer
- Institute of Materials Research, Division for Metallic Biomaterials, Helmholtz-Zentrum Geesthacht (HZG), Geesthacht 21502, Germany
| | | |
Collapse
|
24
|
Lin Z, Shen D, Zhou W, Zheng Y, Kong T, Liu X, Wu S, Chu PK, Zhao Y, Wu J, Cheung KM, Yeung KW. Regulation of extracellular bioactive cations in bone tissue microenvironment induces favorable osteoimmune conditions to accelerate in situ bone regeneration. Bioact Mater 2021; 6:2315-2330. [PMID: 33553818 PMCID: PMC7840811 DOI: 10.1016/j.bioactmat.2021.01.018] [Citation(s) in RCA: 76] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Revised: 01/14/2021] [Accepted: 01/16/2021] [Indexed: 12/25/2022] Open
Abstract
The design of orthopedic biomaterials has gradually shifted from “immune-friendly” to “immunomodulatory,” in which the biomaterials are able to modulate the inflammatory response via macrophage polarization in a local immune microenvironment that favors osteogenesis and implant-to-bone osseointegration. Despite the well-known effects of bioactive metallic ions on osteogenesis, how extracellular metallic ions manipulate immune cells in bone tissue microenvironments toward osteogenesis and subsequent bone formation has rarely been studied. Herein, we investigate the osteoimmunomodulatory effect of an extracellular bioactive cation (Mg2+) in the bone tissue microenvironment using custom-made poly lactic-co-glycolic acid (PLGA)/MgO-alendronate microspheres that endow controllable release of magnesium ions. The results suggest that the Mg2+-controlled tissue microenvironment can effectively induce macrophage polarization from the M0 to M2 phenotype via the enhancement of anti-inflammatory (IL-10) and pro-osteogenic (BMP-2 and TGF-β1) cytokines production. It also generates a favorable osteoimmune microenvironment that facilitates the proliferation and osteogenic differentiation of bone marrow mesenchymal stem cells. The in vivo results further verify that a large amount of bony tissue, with comparable bone mineral density and mechanical properties, has been generated at an early post-surgical stage in rat intramedullary bone defect models. This study demonstrates that the concept of in situ immunomodulated osteogenesis can be realized in a controlled magnesium tissue microenvironment. Controlled release of Mg2+ was achieved by the microfluidic electrosprayed PLGA/MgO-alendronate microspheres. PLGA/MgO-alendronate microspheres with satisfied osteoimmunomodulatory properties accelerated bone regeneration in vivo. Extracellular Mg2+ induced macrophage switch to M2 phenotype with enhanced expressions of IL-10, BMP-2 and TGF-β1. Osteoimmune microenvironment favourably upregulated the osteogenic expressions of BMSCs.
Collapse
Affiliation(s)
- Zhengjie Lin
- College of Chemistry and Environmental Engineering, Shenzhen University, Shenzhen, 518060, China
- Department of Orthopaedics and Traumatology, The University of Hong Kong, Hong Kong, China
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096, China
- Corresponding author. College of Chemistry and Environmental Engineering, Shenzhen University, China.
| | - Danni Shen
- Department of Materials Science and Engineering, College of Engineering, Peking University, Beijing, 100871, China
- Shenzhen Key Laboratory for Innovative Technology in Orthopaedic Trauma, The University of Hong Kong Shenzhen Hospital, 1 Haiyuan 1st Road, Futian District, Shenzhen, China
| | - Weixiao Zhou
- Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, Department of Biomedical Engineering, School of Medicine, Shenzhen University, Shenzhen, 518060, China
| | - Yufeng Zheng
- Department of Materials Science and Engineering, College of Engineering, Peking University, Beijing, 100871, China
| | - Tiantian Kong
- Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, Department of Biomedical Engineering, School of Medicine, Shenzhen University, Shenzhen, 518060, China
| | - Xuanyong Liu
- State Key Laboratory of High Performance Ceramics and Superfine Microstructure, Shanghai Institute of Ceramics, Chinese Academy of Sciences, Shanghai, 200050, China
| | - Shuilin Wu
- School of Materials Science & Engineering, the Key Laboratory of Advanced Ceramics and Machining Technology by the Ministry of Education of China, Tianjin University, Tianjin, 300072, China
| | - Paul K. Chu
- Department of Physics, Department of Materials Science and Engineering, City University of Hong Kong, Tat Chee Avenue, Kowloon, Hong Kong, China
| | - Ying Zhao
- Centre for Human Tissues and Organs Degeneration, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
| | - Jun Wu
- Shenzhen Key Laboratory for Innovative Technology in Orthopaedic Trauma, The University of Hong Kong Shenzhen Hospital, 1 Haiyuan 1st Road, Futian District, Shenzhen, China
| | - Kenneth M.C. Cheung
- Department of Orthopaedics and Traumatology, The University of Hong Kong, Hong Kong, China
| | - Kelvin W.K. Yeung
- Department of Orthopaedics and Traumatology, The University of Hong Kong, Hong Kong, China
- Shenzhen Key Laboratory for Innovative Technology in Orthopaedic Trauma, The University of Hong Kong Shenzhen Hospital, 1 Haiyuan 1st Road, Futian District, Shenzhen, China
- Corresponding author. Department of Orthopaedics and Traumatology, The University of Hong Kong, Hong Kong, China.
| |
Collapse
|
25
|
Tang CF, Ding H, Jiao RQ, Wu XX, Kong LD. Possibility of magnesium supplementation for supportive treatment in patients with COVID-19. Eur J Pharmacol 2020; 886:173546. [PMID: 32931782 PMCID: PMC7486870 DOI: 10.1016/j.ejphar.2020.173546] [Citation(s) in RCA: 67] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Revised: 07/30/2020] [Accepted: 09/08/2020] [Indexed: 12/16/2022]
Abstract
Magnesium as an enzymatic activator is essential for various physiological functions such as cell cycle, metabolic regulation, muscle contraction, and vasomotor tone. A growing body of evidence supports that magnesium supplementation (mainly magnesium sulfate and magnesium oxide) prevents or treats various types of disorders or diseases related to respiratory system, reproductive system, nervous system, digestive system, and cardiovascular system as well as kidney injury, diabetes and cancer. The ongoing pandemic coronavirus disease 19 (COVID-19) characterized by respiratory tract symptoms with different degrees of important organ and tissue damages has attracted global attention. Particularly, effective drugs are still lacking in the COVID-19 therapy. In this review, we find and summarize the effectiveness of magnesium supplementation on the disorders or diseases, and provide a reference to the possibility of magnesium supplementation for supportive treatment in patients with COVID-19.
Collapse
Affiliation(s)
- Chuan-Feng Tang
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, 210023, People's Republic of China
| | - Hong Ding
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, 210023, People's Republic of China
| | - Rui-Qing Jiao
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, 210023, People's Republic of China
| | - Xing-Xin Wu
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, 210023, People's Republic of China
| | - Ling-Dong Kong
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, 210023, People's Republic of China.
| |
Collapse
|
26
|
Liu M, Dudley SC. Magnesium, Oxidative Stress, Inflammation, and Cardiovascular Disease. Antioxidants (Basel) 2020; 9:E907. [PMID: 32977544 PMCID: PMC7598282 DOI: 10.3390/antiox9100907] [Citation(s) in RCA: 74] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Revised: 09/17/2020] [Accepted: 09/18/2020] [Indexed: 12/15/2022] Open
Abstract
Hypomagnesemia is commonly observed in heart failure, diabetes mellitus, hypertension, and cardiovascular diseases. Low serum magnesium (Mg) is a predictor for cardiovascular and all-cause mortality and treating Mg deficiency may help prevent cardiovascular disease. In this review, we discuss the possible mechanisms by which Mg deficiency plays detrimental roles in cardiovascular diseases and review the results of clinical trials of Mg supplementation for heart failure, arrhythmias and other cardiovascular diseases.
Collapse
Affiliation(s)
- Man Liu
- Division of Cardiology, Department of Medicine, the Lillehei Heart Institute, University of Minnesota at Twin Cities, Minneapolis, MN 55455, USA
| | - Samuel C. Dudley
- Division of Cardiology, Department of Medicine, the Lillehei Heart Institute, University of Minnesota at Twin Cities, Minneapolis, MN 55455, USA
| |
Collapse
|
27
|
Magnesium sulfate prophylaxis attenuates the postpartum effects of preeclampsia by promoting M2 macrophage polarization. Hypertens Res 2020; 44:13-22. [PMID: 32719464 DOI: 10.1038/s41440-020-0511-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2020] [Revised: 06/13/2020] [Accepted: 06/18/2020] [Indexed: 12/12/2022]
Abstract
Preeclampsia is a complex disorder that is characterized by new onset hypertension and proteinuria at or after 20 weeks of gestation. Preeclampsia is a leading cause of maternal and fetal morbidity and mortality. MgSO4 is commonly used to treat severe preeclampsia, but its mechanism of action is poorly understood, and investigations into the effects of MgSO4 during the postpartum period are lacking. In this study, timed-pregnant Sprague-Dawley rats received low-dose lipopolysaccharide (LPS) on gestational day 14 to induce preeclampsia. Maternal and fetal outcomes and the macrophage profile 1 week after delivery were explored. On postpartum day (PD) 7, the maternal systolic blood pressure and urinary protein level were significantly increased, the number of M1 macrophages was increased and the number of M2 macrophages was decreased in the maternal kidney and brain; the median duration of gestation, the number of live fetuses, and the fetal weight/placenta weight ratio were significantly decreased; and the percentage of growth-restricted pups and fetal mortality were significantly increased in preeclampsia rats compared to normal pregnant control rats. Prophylactic MgSO4 decreased blood pressure at PD7, improved pregnancy outcomes, and promoted the polarization of M2 macrophages in the kidney and of M2 microglia in the brain of preeclampsia rats. These findings confirm that the pathophysiology of preeclampsia involves the dysregulation of the inflammatory response and the activation of M1 macrophages in several target organs during pregnancy. MgSO4 prophylaxis attenuates the postpartum effects of preeclampsia by promoting M2 macrophage polarization in the maternal kidney and brain.
Collapse
|
28
|
Analgesic management of uncomplicated acute sickle‐cell pain crisis in pediatrics: a systematic review and meta‐analysis. JORNAL DE PEDIATRIA (VERSÃO EM PORTUGUÊS) 2020. [DOI: 10.1016/j.jpedp.2019.05.024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
|
29
|
Saramba MI, Shakya S, Zhao D. Analgesic management of uncomplicated acute sickle-cell pain crisis in pediatrics: a systematic review and meta-analysis. J Pediatr (Rio J) 2020; 96:142-158. [PMID: 31351033 PMCID: PMC9432155 DOI: 10.1016/j.jped.2019.05.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/26/2019] [Revised: 04/18/2019] [Accepted: 05/08/2019] [Indexed: 01/22/2023] Open
Abstract
OBJECTIVES To capture evidence of the efficacy and safety of pharmacological analgesia for uncomplicated acute sickle-cell pain in pediatric patients compared to placebo. SOURCES OF DATA Searches for key evidence were performed from March 1 to 31, 2018, for randomized controlled trials of pharmacological analgesia compared to placebo for uncomplicated acute sickle-cell pain in a pediatric sample. The authors searched ten scientific databases including, among others, PubMed, MEDLINE, Embase, and Clinicaltrials.gov for this systematic review and meta-analysis. SUMMARY OF THE FINDINGS Four trials (n=227) were selected by the inclusion criteria (intranasal fentanyl, intravenous magnesium, arginine, and inhaled nitric oxide). The quality of evidence ranged from low to moderate for each outcome. Meta-analysis of changes in the ladder of pain score (p=0.72), length-of-stay in hospital (p=0.65), and amount of narcotics used during the study (p=0.10) showed non-statistically significant differences and a lack of amelioration provided by pharmaceutical analgesics in treatment group. The adverse events reported that more participants in the intervention arm underwent pain, with statistically significant differences at the drug delivery site in studies using intranasal fentanyl and intravenous magnesium (p=0.03). CONCLUSIONS Pharmacological analgesia appears to be uncertain in improving the intensity and providing relief of acute pain crisis in pediatric patients with sickle-cell anemia. With respect to clinical advantage, no decisive deduction about the clinical efficacy may be made regarding these medications in acute sickle-cell pain management in the pediatric age group.
Collapse
Affiliation(s)
- Manou Irmina Saramba
- Zhongnan Hospital of Wuhan University, Children's Digital and Health Data Research Center, Department of Pediatrics, Wuhan, China
| | - Sandeep Shakya
- Zhongnan Hospital of Wuhan University, Children's Digital and Health Data Research Center, Department of Pediatrics, Wuhan, China
| | - Dongchi Zhao
- Zhongnan Hospital of Wuhan University, Children's Digital and Health Data Research Center, Department of Pediatrics, Wuhan, China.
| |
Collapse
|
30
|
Ozen M, Xie H, Shin N, Al Yousif G, Clemens J, McLane MW, Lei J, Burd I. Magnesium sulfate inhibits inflammation through P2X7 receptors in human umbilical vein endothelial cells. Pediatr Res 2020; 87:463-471. [PMID: 31493768 PMCID: PMC7035964 DOI: 10.1038/s41390-019-0557-7] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/18/2018] [Revised: 08/09/2019] [Accepted: 08/23/2019] [Indexed: 01/02/2023]
Abstract
BACKGROUND Magnesium sulfate (MgSO4) is utilized for fetal neuroprotection in preterm birth but its mechanism of action is still poorly understood. P2X7 receptor (P2X7R) is required for secretion of IL-1β, and can be blocked by divalent cations such as magnesium (Mg) and its own antagonist, Brilliant Blue G (BBG). We sought to determine whether during inflammation MgSO4 can block endothelial IL-1β secretion, using an in-vitro model. METHODS Human umbilical vein endothelial cell (HUVEC) cultures were treated with varying doses of LPS, 2'(3)-Ο-(4-Benzoylbenzoyl) adenosine-5'-triphosphate (BzATP), BBG and MgSO4 for 3- or 24 h. We determined cell cytotoxicity, apoptosis, IL-1β mRNA expression, IL-1β production and secretion and P2X7R expression on HUVECs. RESULTS We demonstrated that MgSO4 is efficacious in blocking IL-1β-mediated-inflammation in HUVECs, at both the initiation and propagation phases of inflammation. MgSO4 exerts these anti-inflammatory effects via downregulation of P2X7Rs on HUVECs. CONCLUSION LPS-exposure increases IL-1β production and secretion in HUVECs, which is further intensified by P2X7R agonist, BzATP while MgSO4 inhibits IL-1β in both presence and absence of BzATP. This effect is similar to the results of P2X7R antagonist, BBG, suggesting that the anti-inflammatory effects of MgSO4 is through P2X7R.
Collapse
Affiliation(s)
- Maide Ozen
- Integrated Research Center for Fetal Medicine, Department of Gynecology and Obstetrics, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA.
- Department of Pediatrics, Neonatal-Perinatal Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA.
| | - Han Xie
- Integrated Research Center for Fetal Medicine, Department of Gynecology and Obstetrics, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Na Shin
- Integrated Research Center for Fetal Medicine, Department of Gynecology and Obstetrics, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Ghada Al Yousif
- Integrated Research Center for Fetal Medicine, Department of Gynecology and Obstetrics, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Julia Clemens
- Integrated Research Center for Fetal Medicine, Department of Gynecology and Obstetrics, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Michael W McLane
- Integrated Research Center for Fetal Medicine, Department of Gynecology and Obstetrics, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Jun Lei
- Integrated Research Center for Fetal Medicine, Department of Gynecology and Obstetrics, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Irina Burd
- Integrated Research Center for Fetal Medicine, Department of Gynecology and Obstetrics, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| |
Collapse
|
31
|
Liu F, Ling Z, Tang C, Yi F, Chen YQ. Moderation effects of food intake on the relationship between urinary microbiota and urinary interleukin-8 in female type 2 diabetic patients. PeerJ 2020; 8:e8481. [PMID: 32025384 PMCID: PMC6993747 DOI: 10.7717/peerj.8481] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2019] [Accepted: 12/27/2019] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND Our previous study demonstrated that the composition of the urinary microbiota in female patients with type 2 diabetes mellitus (T2DM) was correlated with the concentration of urinary interleukin (IL)-8. As the composition of urine is mainly determined by diet, diet might mediate the correlation. METHODS Seventy female T2DM patients and 70 healthy controls (HCs) were recruited. Midstream urine was used for the urine specimens. Urinary IL-8 was determined by enzyme-linked immunosorbent assay. A Chinese Food Frequency Questionnaire was used to collect food intake data. The independent variables in the hierarchical regression analysis were the relative abundances of the bacterial genera and species that were significantly different between the T2DM and HCs and between the T2DM patients with and without detectable urinary IL-8, and the bacterial genera associated with IL-8 concentration in the multiple regression model reported in our previous research. IL-8 concentration was the dependent variable, and nutrient intakes were moderator variables. RESULTS Fiber and vitamin B3 and E intake exerted enhancing effects, and water intake exerted a buffering effect, on the positive relationship between the relative abundance of Ruminococcus and IL-8 concentration (p < 0.05). Cholesterol and magnesium intake exerted enhancing effects on the positive relationship between the relative abundance of Comamonas and IL-8 concentration (p < 0.05). CONCLUSION Modulating T2DM patients' dietary patterns may prevent bladder inflammation.
Collapse
Affiliation(s)
- Fengping Liu
- Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu, China
| | - Zongxin Ling
- Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Chulei Tang
- Xiangya Nursing School, Central South University, Changsha, China
| | - Fendi Yi
- Endocrinology Department, The Affiliated Yancheng Hospital of Southeast University Medical College, Yancheng, China
| | - Yong Q. Chen
- Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu, China
| |
Collapse
|
32
|
Jin L, Chen C, Li Y, Yuan F, Gong R, Wu J, Zhang H, Kang B, Yuan G, Zeng H, Chen T. A Biodegradable Mg-Based Alloy Inhibited the Inflammatory Response of THP-1 Cell-Derived Macrophages Through the TRPM7-PI3K-AKT1 Signaling Axis. Front Immunol 2019; 10:2798. [PMID: 31849975 PMCID: PMC6902094 DOI: 10.3389/fimmu.2019.02798] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2019] [Accepted: 11/14/2019] [Indexed: 12/16/2022] Open
Abstract
Mg-based alloys might be ideal biomaterials in clinical applications owing to favorable mechanical properties, biodegradability, biocompatibility, and especially their anti-inflammatory properties. However, the precise signaling mechanism underlying the inhibition of inflammation by Mg-based alloys has not been elucidated. Here, we investigated the effects of a Mg-2.1Nd-0.2Zn-0.5Zr alloy (denoted as JDBM) on lipopolysaccharide (LPS)-induced macrophages. THP-1 cell-derived macrophages were cultured on JDBM, Ti-6Al-4V alloy (Ti), 15% extract of JDBM, and 7.5 mM of MgCl2 for 1 h before the addition of LPS for an indicated time; the experiments included negative and positive controls. Our results showed JDBM, extract, and MgCl2 could decrease LPS-induced tumor necrosis factor (TNF) and interleukin (IL)-6 expression. However, there were no morphologic changes in macrophages on Ti or JDBM. Mechanically, extract and MgCl2 downregulated the expression of toll-like receptor (TLR)-4 and MYD88 compared with the positive control and inhibited LPS-induced nuclear factor-kappa B (NF-κB) and mitogen-activated protein kinase (MAPK) signaling pathways by inactivation of the phosphorylation of IKK-α/β, IKβ-α, P65, P38, and JNK. Additionally, the LPS-induced reactive oxygen species (ROS) expression was also decreased by extract and MgCl2. Interestingly, the expression of LPS-induced TNF and IL-6 could be recovered by knocking down TRPM7 of macrophages, in the presence of extract or MgCl2. Mechanically, the activities of AKT and AKT1 were increased by extract or MgCl2 with LPS and were blocked by a PI3K inhibitor, whereas siRNA TRPM7 inhibited only AKT1. Together, our results demonstrated the degradation products of Mg-based alloy, especially magnesium, and resolved inflammation by activation of the TRPM7-PI3K-AKT1 signaling pathway, which may be a potential advantage or target to promote biodegradable Mg-based alloy applications.
Collapse
Affiliation(s)
- Liang Jin
- School of Biomedical Engineering, Med-X Research Institute, Shanghai Jiao Tong University, Shanghai, China
- State Key Laboratory of Metal Matrix Composite, National Engineering Research Center of Light Alloy Net Forming, Shanghai Jiao Tong University, Shanghai, China
- Division of Immunology, Shanghai Children's Medical Center, Institute of Pediatric Translational Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Chenxin Chen
- State Key Laboratory of Metal Matrix Composite, National Engineering Research Center of Light Alloy Net Forming, Shanghai Jiao Tong University, Shanghai, China
| | - Yutong Li
- State Key Laboratory of Metal Matrix Composite, National Engineering Research Center of Light Alloy Net Forming, Shanghai Jiao Tong University, Shanghai, China
| | - Feng Yuan
- State Key Laboratory of Metal Matrix Composite, National Engineering Research Center of Light Alloy Net Forming, Shanghai Jiao Tong University, Shanghai, China
| | - Ruolan Gong
- Division of Immunology, Shanghai Children's Medical Center, Institute of Pediatric Translational Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Department of Allergy and Immunology, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jing Wu
- Division of Immunology, Shanghai Children's Medical Center, Institute of Pediatric Translational Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Hua Zhang
- State Key Laboratory of Metal Matrix Composite, National Engineering Research Center of Light Alloy Net Forming, Shanghai Jiao Tong University, Shanghai, China
| | - Bin Kang
- Department of Orthopaedics, Peking University Shenzhen Hospital of Medicine, Shenzhen, China
| | - Guangyin Yuan
- School of Biomedical Engineering, Med-X Research Institute, Shanghai Jiao Tong University, Shanghai, China
- State Key Laboratory of Metal Matrix Composite, National Engineering Research Center of Light Alloy Net Forming, Shanghai Jiao Tong University, Shanghai, China
| | - Hui Zeng
- Department of Orthopaedics, Peking University Shenzhen Hospital of Medicine, Shenzhen, China
| | - Tongxin Chen
- Division of Immunology, Shanghai Children's Medical Center, Institute of Pediatric Translational Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Department of Allergy and Immunology, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
33
|
Wu H, Yin Y, Hu X, Peng C, Liu Y, Li Q, Huang W, Huang Q. Effects of Environmental pH on Macrophage Polarization and Osteoimmunomodulation. ACS Biomater Sci Eng 2019; 5:5548-5557. [DOI: 10.1021/acsbiomaterials.9b01181] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
- Hong Wu
- State Key Laboratory of Powder Metallurgy, Central South University, Changsha 410083, China
- Shenzhen Zhong Jin Ling Nan Nonfemet Co., Ltd, Shenzhen 518040, China
- School of Materials Science and Engineering, Northwestern Polytechnical University, Xi’an 710072, China
| | - Yong Yin
- State Key Laboratory of Powder Metallurgy, Central South University, Changsha 410083, China
| | - Xiaobo Hu
- The First Department of Breast Surgery, Hunan Cancer Hospital, Changsha 410013, China
| | - Cheng Peng
- The Third Xiangya Hospital, Central South University, Changsha 410013, China
| | - Yong Liu
- State Key Laboratory of Powder Metallurgy, Central South University, Changsha 410083, China
| | - Qingxiang Li
- Shenzhen Zhong Jin Ling Nan Nonfemet Co., Ltd, Shenzhen 518040, China
| | - Weidong Huang
- School of Materials Science and Engineering, Northwestern Polytechnical University, Xi’an 710072, China
| | - Qianli Huang
- State Key Laboratory of Powder Metallurgy, Central South University, Changsha 410083, China
| |
Collapse
|
34
|
Zhu D, You J, Zhao N, Xu H. Magnesium Regulates Endothelial Barrier Functions through TRPM7, MagT1, and S1P1. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2019; 6:1901166. [PMID: 31559137 PMCID: PMC6755513 DOI: 10.1002/advs.201901166] [Citation(s) in RCA: 51] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/16/2019] [Revised: 06/24/2019] [Indexed: 05/27/2023]
Abstract
Mg2+-deficiency is linked to hypertension, Alzheimer's disease, stroke, migraine headaches, cardiovascular diseases, and diabetes, etc., but its exact role in these pathophysiological conditions remains elusive. Mg2+ can regulate vascular functions, yet the mechanistic insight remains ill-defined. Data show that extracellular Mg2+ enters endothelium mainly through the TRPM7 channel and MagT1 transporter. Mg2+ can act as an antagonist to reduce Ca2+ signaling in endothelium. Mg2+ also reduces the intracellular reactive oxygen species (ROS) level and inflammation. In addition, Mg2+-signaling increases endothelial survival and growth, adhesion, and migration. Endothelial barrier integrity is significantly enhanced with Mg2+-treatment through S1P1-Rac1 pathways and barrier-stabilizing mediators including cAMP, FGF1/2, and eNOS. Mg2+ also promotes cytoskeletal reorganization and junction proteins to tighten up the barrier. Moreover, Mg2+-deficiency enhances endothelial barrier permeability in mice, and Mg2+-treatment rescues histamine-induced transient vessel hyper-permeability in vivo. In summary, Mg2+-deficiency can cause deleterious effects in endothelium integrity, and Mg2+-treatment may be effective in the prevention or treatment of vascular dysfunction.
Collapse
Affiliation(s)
- Donghui Zhu
- Department of Biomedical EngineeringInstitute for Engineering‐Driven MedicineCollege of Engineering and Applied SciencesRenaissance School of MedicineStony Brook UniversityStony BrookNY11794USA
| | - Jing You
- Department of Biomedical EngineeringUniversity of North TexasDentonTX76207USA
| | - Nan Zhao
- Department of Biomedical EngineeringPennsylvania State UniversityState CollegePA16802USA
| | - Huaxi Xu
- Sanford Burnham Prebys Medical Discovery InstituteLa JollaCA92037USA
| |
Collapse
|
35
|
Magnesium Sulfate Mitigates the Progression of Monocrotaline Pulmonary Hypertension in Rats. Int J Mol Sci 2019; 20:ijms20184622. [PMID: 31540416 PMCID: PMC6770589 DOI: 10.3390/ijms20184622] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2019] [Revised: 09/13/2019] [Accepted: 09/16/2019] [Indexed: 12/16/2022] Open
Abstract
We investigated whether magnesium sulfate (MgSO4) mitigated pulmonary hypertension progression in rats. Pulmonary hypertension was induced by a single intraperitoneal injection of monocrotaline (60 mg/kg). MgSO4 (100 mg/kg) was intraperitoneally administered daily for 3 weeks, from the seventh day after monocrotaline injection. Adult male rats were randomized into monocrotaline (MCT) or monocrotaline plus MgSO4 (MM) groups (n = 15 per group); control groups were maintained simultaneously. For analysis, surviving rats were euthanized on the 28th day after receiving monocrotaline. The survival rate was higher in the MM group than in the MCT group (100% versus 73.3%, p = 0.043). Levels of pulmonary artery wall thickening, α-smooth muscle actin upregulation, right ventricular systolic pressure increase, and right ventricular hypertrophy were lower in the MM group than in the MCT group (all p < 0.05). Levels of lipid peroxidation, mitochondrial injury, inflammasomes and cytokine upregulation, and apoptosis in the lungs and right ventricle were lower in the MM group than in the MCT group (all p < 0.05). Notably, the mitigation effects of MgSO4 on pulmonary artery wall thickening and right ventricular hypertrophy were counteracted by exogenous calcium chloride. In conclusion, MgSO4 mitigates pulmonary hypertension progression, possibly by antagonizing calcium.
Collapse
|
36
|
Dysregulation of inflammatory cytokines and inhibition of VEGFA in the human umbilical cord are associated with negative pregnancy outcomes. Placenta 2019; 87:16-22. [PMID: 31525711 DOI: 10.1016/j.placenta.2019.09.002] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/17/2019] [Revised: 08/17/2019] [Accepted: 09/02/2019] [Indexed: 11/22/2022]
Abstract
INTRODUCTION Cytokines and vascular endothelial growth factors (VEGF) are involved in all aspects of pregnancy: from placentation, through fetal development, parturition and neonatal well-being. Umbilical cord inflammatory cytokines and/or VEGF have not been well studied with respect to dysregulation associated with disorders of pregnancy or maternal/neonatal outcomes. METHODS Here we have used multiplex ELISA to screen umbilical cord lysates (comprising cord blood, endothelia and Wharton's jelly, n = 380), for levels of IFN-γ, IL1-β, IL-6, IL-8, IL-10, TNF-α and VEGFs A, C and D and associations with 46 ICD9/10 codes encompassing obstetric, maternal and neonatal variables. RESULTS No significant differences were observed for IFNγ, VEGFC or VEGFD with any clinical outcomes. The cytokines IL1-β, IL-6, IL-8, IL-10, and TNF-α showed varying levels of induction and suppression with primarily fetal-placental and neonatal complications. The largest number of significant differences between umbilical cytokines and clinical outcomes were observed for chorioamnionitis (IL1-β, IL-6, IL-8, TNF-α), and meconium passage during birth (IL1-β, IL-6, IL-8) where significant pro-inflammatory responses occurred and sex differences in IL-8 expression were noted. In contrast, gonococcal infection showed suppressed immune response significantly lowering IL1-β, IL-6, IL-8, IL-10 and TNF-α. For 12/46 negative pregnancy outcomes, strong suppression of VEGFA occurred. DISCUSSION Angiogenic and inflammatory changes in the umbilical cord could be detrimental by increasing vascular permeability in the umbilical artery or vein and/or altering vascular tone, either of which would alter blood flow affecting delivery and removal of compounds. Further elucidation of inflammatory responses in the umbilical cord may provide mechanistic understanding of adverse pregnancy outcomes.
Collapse
|
37
|
Shimohata H, Yamashita M, Ohgi K, Tsujimoto R, Maruyama H, Takayasu M, Hirayama K, Kobayashi M. The relationship between serum magnesium levels and mortality in non-diabetic hemodialysis patients: A 10-year follow-up study. Hemodial Int 2019; 23:369-374. [PMID: 31037843 DOI: 10.1111/hdi.12759] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2018] [Revised: 03/15/2019] [Accepted: 04/12/2019] [Indexed: 11/28/2022]
Abstract
Introduction Recently, although there are many reports showing that serum magnesium concentration is a predictor of mortality in dialysis patients, the observation periods of those reports were of short duration, typically around 12 months. Thus, we investigated this relationship over a longer follow-up period. Methods This retrospective, observational study included a total of 83 non-diabetic hemodialysis patients. The follow-up period was 120 months. Patients were divided into two groups, those with serum magnesium ≥2.5 mg/dL (Mg ≥2.5 mg/dL group) and serum magnesium <2.5 mg/dL (Mg <2.5 mg/dL group), and Kaplan-Meier analysis and Cox proportional hazards analysis were conducted. In addition to the above analysis, single and multiple regression analysis were performed at baseline to reveal the relationship between serum magnesium and clinical parameters. Findings During the follow-up period, 31 out of 83 patients died. Kaplan-Meier analysis showed a significantly higher incidence of death in the Mg <2.5 mg/dL group (log-rank test 4.951, P = 0.026). Multivariate Cox proportional hazards analysis showed a 62% decreased risk of mortality in the Mg ≥2.5 mg/dL group compared to the Mg <2.5 mg/dL group after adjustment for several confounding factors. Simple correlation coefficient analysis showed positive correlations of serum magnesium levels with serum creatinine, phosphorus, high-density lipoprotein, ankle-brachial index and KT/V, and a negative correlation with age. Multiple linear regression analysis showed that the ankle-brachial index was the only parameter that had a positive and significant correlation with the serum magnesium level. Conclusion Our study demonstrated that higher serum magnesium levels were associated with improved survival in non-diabetic hemodialysis patients.
Collapse
Affiliation(s)
- Homare Shimohata
- Department of Nephrology, Tokyo Medical University Ibaraki Medical Center, Ibaraki, Japan
| | - Marina Yamashita
- Department of Nephrology, Tokyo Medical University Ibaraki Medical Center, Ibaraki, Japan
| | - Kentaro Ohgi
- Department of Nephrology, Tokyo Medical University Ibaraki Medical Center, Ibaraki, Japan
| | - Ryuji Tsujimoto
- Department of Nephrology, Tokyo Medical University Ibaraki Medical Center, Ibaraki, Japan
| | - Hiroshi Maruyama
- Department of Nephrology, Tokyo Medical University Ibaraki Medical Center, Ibaraki, Japan
| | - Mamiko Takayasu
- Department of Nephrology, Tokyo Medical University Ibaraki Medical Center, Ibaraki, Japan
| | - Kouichi Hirayama
- Department of Nephrology, Tokyo Medical University Ibaraki Medical Center, Ibaraki, Japan
| | - Masaki Kobayashi
- Department of Nephrology, Tokyo Medical University Ibaraki Medical Center, Ibaraki, Japan
| |
Collapse
|
38
|
Jia B, Zong L, Lee JY, Lei J, Zhu Y, Xie H, Clemens JL, Feller MC, Na Q, Dong J, McLane MW, Jones-Beatty K, Burd I. Maternal Supplementation of Low Dose Fluoride Alleviates Adverse Perinatal Outcomes Following Exposure to Intrauterine Inflammation. Sci Rep 2019; 9:2575. [PMID: 30796233 PMCID: PMC6385257 DOI: 10.1038/s41598-018-38241-8] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2018] [Accepted: 12/12/2018] [Indexed: 11/09/2022] Open
Abstract
Maternal periodontal disease has been linked to adverse pregnancy sequelae, including preterm birth (PTB); yet, root planing and scaling in pregnancy has not been associated with improved perinatal outcomes. Fluoride, a cariostatic agent, has been added to drinking water and dental products to prevent caries and improve dental health. The objective of this study was to explore the effects of fluoride supplementation using a mouse model of preterm birth and perinatal sequalae. Pregnant mice were fed low dose fluoride (LF-) or high dose fluoride (HF-) and given intrauterine injections of lipopolysaccharide (LPS) or phosphate-buffered saline (PBS). We found that LPS + LF- significantly increased livebirths, pup survival, and litter size compared to LPS alone. Moreover, offspring from the LPS + LF- group exhibited significantly improved neuromotor performance and more neurons compared to those from the LPS group. Additionally, LF- treatment on human umbilical vein endothelial cells (HUVECs) increased cell viability and decreased oxidative stress after treatment with LPS. Collectively, our data demonstrates that maternal LF- supplementation during pregnancy postpones the onset of PTB, acts to increase the liveborn rate and survival time of newborns, and reduces perinatal brain injury in cases of intrauterine inflammation.
Collapse
Affiliation(s)
- Bei Jia
- Integrated Research Center for Fetal Medicine, Division of Maternal Fetal Medicine, Department of Gynecology and Obstetrics, Johns Hopkins University, School of Medicine, Baltimore, MD, USA
- The Center for Prenatal and Hereditary Disease Diagnosis, Department of Obstetrics and Gynecology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Lu Zong
- Integrated Research Center for Fetal Medicine, Division of Maternal Fetal Medicine, Department of Gynecology and Obstetrics, Johns Hopkins University, School of Medicine, Baltimore, MD, USA
| | - Ji Yeon Lee
- Integrated Research Center for Fetal Medicine, Division of Maternal Fetal Medicine, Department of Gynecology and Obstetrics, Johns Hopkins University, School of Medicine, Baltimore, MD, USA
| | - Jun Lei
- Integrated Research Center for Fetal Medicine, Division of Maternal Fetal Medicine, Department of Gynecology and Obstetrics, Johns Hopkins University, School of Medicine, Baltimore, MD, USA
| | - Yan Zhu
- Integrated Research Center for Fetal Medicine, Division of Maternal Fetal Medicine, Department of Gynecology and Obstetrics, Johns Hopkins University, School of Medicine, Baltimore, MD, USA
| | - Han Xie
- Integrated Research Center for Fetal Medicine, Division of Maternal Fetal Medicine, Department of Gynecology and Obstetrics, Johns Hopkins University, School of Medicine, Baltimore, MD, USA
| | - Julia L Clemens
- Integrated Research Center for Fetal Medicine, Division of Maternal Fetal Medicine, Department of Gynecology and Obstetrics, Johns Hopkins University, School of Medicine, Baltimore, MD, USA
| | - Mia C Feller
- Integrated Research Center for Fetal Medicine, Division of Maternal Fetal Medicine, Department of Gynecology and Obstetrics, Johns Hopkins University, School of Medicine, Baltimore, MD, USA
| | - Quan Na
- Integrated Research Center for Fetal Medicine, Division of Maternal Fetal Medicine, Department of Gynecology and Obstetrics, Johns Hopkins University, School of Medicine, Baltimore, MD, USA
| | - Jie Dong
- Integrated Research Center for Fetal Medicine, Division of Maternal Fetal Medicine, Department of Gynecology and Obstetrics, Johns Hopkins University, School of Medicine, Baltimore, MD, USA
| | - Michael W McLane
- Integrated Research Center for Fetal Medicine, Division of Maternal Fetal Medicine, Department of Gynecology and Obstetrics, Johns Hopkins University, School of Medicine, Baltimore, MD, USA
| | - Kimberly Jones-Beatty
- Integrated Research Center for Fetal Medicine, Division of Maternal Fetal Medicine, Department of Gynecology and Obstetrics, Johns Hopkins University, School of Medicine, Baltimore, MD, USA
| | - Irina Burd
- Integrated Research Center for Fetal Medicine, Division of Maternal Fetal Medicine, Department of Gynecology and Obstetrics, Johns Hopkins University, School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
39
|
Hu T, Xu H, Wang C, Qin H, An Z. Magnesium enhances the chondrogenic differentiation of mesenchymal stem cells by inhibiting activated macrophage-induced inflammation. Sci Rep 2018; 8:3406. [PMID: 29467509 PMCID: PMC5821731 DOI: 10.1038/s41598-018-21783-2] [Citation(s) in RCA: 77] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2017] [Accepted: 02/09/2018] [Indexed: 12/22/2022] Open
Abstract
Magnesium deficiency increases the generation of pro-inflammatory cytokines, which is consistently accompanied by the sensitization of cells such as neutrophils, macrophages and endothelial cells. We investigated the potential of magnesium to regulate macrophage polarization and macrophage-induced inflammation with or without lipopolysaccharide (LPS) and interferon-γ (IFN-γ) activation and further elucidated whether these effects impact the inhibitory functions of activated macrophage-induced inflammation on cartilage regeneration. The results showed that magnesium inhibited the activation of macrophages, as indicated by a significant reduction in the percentage of CCR7-positive cells, while the percentage of CD206-positive cells decreased to a lesser degree. After activation, both pro-inflammatory and anti-inflammatory cytokines were down-regulated at the mRNA level and certain cytokines (IL-1β, IL-6 and IL-10) were decreased in the cell supernatant with the addition of magnesium. Moreover, magnesium decreased the nuclear translocation and phosphorylation of nuclear factor-κB (NF-κB) to impede its activation. A modified micromass culture system was applied to assess the effects of activated macrophage-conditioned medium with or without magnesium treatment on the chondrogenic differentiation of human bone marrow mesenchymal stem cells (hBMSCs). Magnesium enhanced the chondrogenic differentiation of hBMSCs by reversing the adverse effects of activated macrophage-induced inflammation.
Collapse
Affiliation(s)
- Tu Hu
- Department of Orthopedics, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Haitao Xu
- Trauma Center, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Chongyang Wang
- Department of Orthopedics, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Hui Qin
- Department of Orthopedics, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Zhiquan An
- Department of Orthopedics, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China.
| |
Collapse
|
40
|
Brenner M, Laragione T, Gulko PS. Short-term low-magnesium diet reduces autoimmune arthritis severity and synovial tissue gene expression. Physiol Genomics 2017; 49:238-242. [DOI: 10.1152/physiolgenomics.00003.2017] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2017] [Revised: 02/13/2017] [Accepted: 02/27/2017] [Indexed: 01/10/2023] Open
Abstract
Magnesium has been suggested to have anti-inflammatory properties in short-term, mostly in vitro studies. To examine the effect of dietary magnesium modifications in arthritis severity and joint damage DA rats were placed on one of three diet regimens before the induction of autoimmune pristane-induced arthritis (PIA): a 4 wk low-magnesium diet, normal diet, and a magnesium-supplemented diet. The diets were switched to a normal diet 14 days after the induction of PIA (typical time of disease onset). Arthritis severity was scored for 38 days, and joints were examined by histology and quantitative PCR for proinflammatory genes. Rats on the low-magnesium diet were significantly and reproducibly protected and had 70% lower median arthritis severity score, with preservation of normal joint histology without erosive changes. Rats on the normal or magnesium-supplemented diets were not protected and developed equally severe and erosive disease. While the dietary modifications were at disease onset ( day 14 postinduction), the protective effect of the short-term low-magnesium diet persisted, suggesting a lasting effect on a critical pathogenic pathway. Rats on the low-magnesium diet had significant reduction in synovial tissue expression of IL-6, RORA, and RORC, which are genes required for the development of Th17 T cells. This study revealed a novel role for dietary magnesium in the regulation of autoimmune arthritis and opens new possibilities for the treatment of autoimmune diseases such as rheumatoid arthritis and psoriatic arthritis with short courses of dietary or drug-induced modulations of magnesium levels.
Collapse
Affiliation(s)
- Max Brenner
- The Feinstein Institute for Medical Research, Manhasset, New York; and
| | - Teresina Laragione
- Division of Rheumatology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Pércio S. Gulko
- Division of Rheumatology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York
| |
Collapse
|
41
|
In vitro and in vivo responses of macrophages to magnesium-doped titanium. Sci Rep 2017; 7:42707. [PMID: 28198427 PMCID: PMC5309879 DOI: 10.1038/srep42707] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2016] [Accepted: 01/12/2017] [Indexed: 12/26/2022] Open
Abstract
Modulating immune response to biomaterials through changing macrophage polarization has been proven to be a promising strategy to elicit beneficial outcomes in tissue repair. The objective of this study was to evaluate the response of macrophage polarization to titanium doped with magnesium (0.1~0.35%), which was prepared through the magnesium plasma immersion ion implantation (Mg PIII) technique. The M1/M2 polarization profile of macrophages was investigated using a murine cell line RAW 264.7 in vitro and a murine air pouch model in vivo. Our results demonstrated that the Mg PIII-treated titanium induced a higher percentage of M2 macrophages and higher concentrations of the anti-inflammatory cytokines interleukin (IL)-4 and IL-10. Genes encoding two growth factors, bone morphogenetic protein 2 (BMP2) and vascular endothelial growth factor (VEGF) were up-regulated, thus indicating the ability of the M2 phenotype to promote wound healing. The nuclear factor κB (NF-κB) signalling pathway was down-regulated. In vivo the Mg PIII -treated titanium elicited a similar effect on macrophage polarization and induced thinner fibrous capsule formation and a decrease in infiltrated cells. These results indicate that Mg PIII treatment has the immunomodulatory potential to elicit the pro-healing M2-polarized macrophage phenotype, thus providing new insight into the development of immunomodulatory biomaterials.
Collapse
|
42
|
Usman S, Foo L, Tay J, Bennett PR, Lees C. Use of magnesium sulfate in preterm deliveries for neuroprotection of the neonate. ACTA ACUST UNITED AC 2017. [DOI: 10.1111/tog.12328] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Affiliation(s)
- Sana Usman
- Imperial College of Science, Technology and Medicine; Institute of Reproductive Developmental Biology; Du Cane Road London W12 0NN UK
| | - Lin Foo
- Imperial College of Science, Technology and Medicine; Institute of Reproductive Developmental Biology; Du Cane Road London W12 0NN UK
| | - Jasmine Tay
- Imperial College of Science, Technology and Medicine; Institute of Reproductive Developmental Biology; Du Cane Road London W12 0NN UK
| | - Phillip R Bennett
- Imperial College Healthcare NHS Trust; The Bays, South Wharf Road, St Mary's Hospital London W2 1NY UK
- Institute of Reproductive Developmental Biology; Queen Charlottes and Chelsea Hospital; Du Cane Road London W12 0HS UK
| | - Christoph Lees
- Imperial College Healthcare NHS Trust; The Bays, South Wharf Road, St Mary's Hospital London W2 1NY UK
- Institute of Reproductive Developmental Biology; Queen Charlottes and Chelsea Hospital; Du Cane Road London W12 0HS UK
| |
Collapse
|
43
|
Cipriano AF, Sallee A, Tayoba M, Cortez Alcaraz MC, Lin A, Guan RG, Zhao ZY, Liu H. Cytocompatibility and early inflammatory response of human endothelial cells in direct culture with Mg-Zn-Sr alloys. Acta Biomater 2017; 48:499-520. [PMID: 27746360 PMCID: PMC5873597 DOI: 10.1016/j.actbio.2016.10.020] [Citation(s) in RCA: 61] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2016] [Revised: 10/04/2016] [Accepted: 10/12/2016] [Indexed: 12/18/2022]
Abstract
Crystalline Mg-Zinc (Zn)-Strontium (Sr) ternary alloys consist of elements naturally present in the human body and provide attractive mechanical and biodegradable properties for a variety of biomedical applications. The first objective of this study was to investigate the degradation and cytocompatibility of four Mg-4Zn-xSr alloys (x=0.15, 0.5, 1.0, 1.5wt%; designated as ZSr41A, B, C, and D respectively) in the direct culture with human umbilical vein endothelial cells (HUVEC) in vitro. The second objective was to investigate, for the first time, the early-stage inflammatory response in cultured HUVECs as indicated by the induction of vascular cellular adhesion molecule-1 (VCAM-1). The results showed that the 24-h in vitro degradation of the ZSr41 alloys containing a β-phase with a Zn/Sr at% ratio ∼1.5 was significantly faster than the ZSr41 alloys with Zn/Sr at% ∼1. Additionally, the adhesion density of HUVECs in the direct culture but not in direct contact with the ZSr41 alloys for up to 24h was not adversely affected by the degradation of the alloys. Importantly, neither culture media supplemented with up to 27.6mM Mg2+ ions nor media intentionally adjusted up to alkaline pH 9 induced any detectable adverse effects on HUVEC responses. In contrast, the significantly higher, yet non-cytotoxic, Zn2+ ion concentration from the degradation of ZSr41D alloy was likely the cause for the initially higher VCAM-1 expression on cultured HUVECs. Lastly, analysis of the HUVEC-ZSr41 interface showed near-complete absence of cell adhesion directly on the sample surface, most likely caused by either a high local alkalinity, change in surface topography, and/or surface composition. The direct culture method used in this study was proposed as a valuable tool for studying the design aspects of Zn-containing Mg-based biomaterials in vitro, in order to engineer solutions to address current shortcomings of Mg alloys for vascular device applications. STATEMENT OF SIGNIFICANCE Magnesium (Mg) alloys specifically designed for biodegradable implant applications have been the focus of biomedical research since the early 2000s. Physicochemical properties of Mg alloys make these metallic biomaterials excellent candidates for temporary biodegradable implants in orthopedic and cardiovascular applications. As Mg alloys continue to be investigated for biomedical applications, it is necessary to understand whether Mg-based materials or the alloying elements have the intrinsic ability to direct an immune response to improve implant integration while avoiding cell-biomaterial interactions leading to chronic inflammation and/or foreign body reactions. The present study utilized the direct culture method to investigate for the first time the in vitro transient inflammatory activation of endothelial cells induced by the degradation products of Zn-containing Mg alloys.
Collapse
Affiliation(s)
- Aaron F Cipriano
- Department of Bioengineering, University of California, Riverside, CA 92521, USA; Materials Science & Engineering Program, University of California, Riverside, CA 92521, USA
| | - Amy Sallee
- Department of Bioengineering, University of California, Riverside, CA 92521, USA
| | - Myla Tayoba
- Department of Bioengineering, University of California, Riverside, CA 92521, USA
| | | | - Alan Lin
- Department of Bioengineering, University of California, Riverside, CA 92521, USA
| | - Ren-Guo Guan
- School of Materials and Metallurgy, Northeastern University, Shenyang 110004, China
| | - Zhan-Yong Zhao
- School of Materials and Metallurgy, Northeastern University, Shenyang 110004, China
| | - Huinan Liu
- Department of Bioengineering, University of California, Riverside, CA 92521, USA; Materials Science & Engineering Program, University of California, Riverside, CA 92521, USA; Stem Cell Center, University of California, Riverside, CA 92521, USA; Cell, Molecular and Developmental Biology Program, University of California, Riverside, CA 92521, USA.
| |
Collapse
|
44
|
Williamson RD, McCarthy C, Kenny LC, O’Keeffe GW. Magnesium sulphate prevents lipopolysaccharide-induced cell death in an in vitro model of the human placenta. Pregnancy Hypertens 2016; 6:356-360. [DOI: 10.1016/j.preghy.2016.08.237] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2016] [Revised: 07/11/2016] [Accepted: 08/26/2016] [Indexed: 10/21/2022]
|
45
|
Naidenow J, Hrgovic I, Doll M, Hailemariam-Jahn T, Lang V, Kleemann J, Kippenberger S, Kaufmann R, Zöller N, Meissner M. Peroxisome proliferator-activated receptor (PPAR) α and δ activators induce ICAM-1 expression in quiescent non stimulated endothelial cells. JOURNAL OF INFLAMMATION-LONDON 2016; 13:27. [PMID: 27547125 PMCID: PMC4992265 DOI: 10.1186/s12950-016-0135-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/11/2016] [Accepted: 08/10/2016] [Indexed: 01/29/2023]
Abstract
Background Peroxisome proliferator-activated receptors (PPARs) are ligand-activated transcription factors that are implicated in the regulation of lipid and glucose homeostasis. PPAR agonists have been shown to control inflammatory processes, in part by inhibiting the expression of distinct proinflammatory genes such as vascular cell adhesion molecule-1 (VCAM-1), IL-8, and intercellular adhesion molecule-1 (ICAM-1). ICAM-1 is an important endothelial membrane receptor that facilitates the transmigration of leukocytes across the endothelium. To date, the influence of PPARα and δ activators on the expression of ICAM-1 in non-induced, quiescent endothelial cells has been unclear. Therefore, we examined the effects of various PPARα and δ agonists on the expression of ICAM-1 in non-stimulated primary human endothelial cells. Results We found that PPARα and PPARδ agonists significantly induced ICAM-1 surface, intracellular protein, and mRNA expression in a time and concentration-dependent manner. The PPARδ induced ICAM-1 expression could be paralleled with a significantly increased T-cell adherence to the endothelial cells whereas PPARα failed to do so. Transcriptional activity studies using an ICAM-1 reporter gene constructs revealed that PPARδ, but not PPARα agonists induced gene expression by stimulating ICAM-1 promoter activity via an Sp1 transcription factor binding site and inhibit the binding of the transcription factors Sp1 and Sp3. Furthermore, we performed mRNA stability assays and found that PPARα and PPARδ agonists increased ICAM-1 mRNA stability. Conclusion Therefore, our data provide the first evidence that PPARα and PPARδ agonists induce ICAM-1 expression in non-stimulated endothelial cells via transcriptional and posttranscriptional mechanisms.
Collapse
Affiliation(s)
- Julia Naidenow
- Department of Dermatology, Venereology and Allergology, Johann Wolfgang Goethe-University, Theodor-Stern-Kai 7, D-60590 Frankfurt am Main, Germany
| | - Igor Hrgovic
- Department of Dermatology, Venereology and Allergology, Johann Wolfgang Goethe-University, Theodor-Stern-Kai 7, D-60590 Frankfurt am Main, Germany
| | - Monika Doll
- Department of Dermatology, Venereology and Allergology, Johann Wolfgang Goethe-University, Theodor-Stern-Kai 7, D-60590 Frankfurt am Main, Germany
| | - Tsige Hailemariam-Jahn
- Department of Dermatology, Venereology and Allergology, Johann Wolfgang Goethe-University, Theodor-Stern-Kai 7, D-60590 Frankfurt am Main, Germany
| | - Victoria Lang
- Department of Dermatology, Venereology and Allergology, Johann Wolfgang Goethe-University, Theodor-Stern-Kai 7, D-60590 Frankfurt am Main, Germany
| | - Johannes Kleemann
- Department of Dermatology, Venereology and Allergology, Johann Wolfgang Goethe-University, Theodor-Stern-Kai 7, D-60590 Frankfurt am Main, Germany
| | - Stefan Kippenberger
- Department of Dermatology, Venereology and Allergology, Johann Wolfgang Goethe-University, Theodor-Stern-Kai 7, D-60590 Frankfurt am Main, Germany
| | - Roland Kaufmann
- Department of Dermatology, Venereology and Allergology, Johann Wolfgang Goethe-University, Theodor-Stern-Kai 7, D-60590 Frankfurt am Main, Germany
| | - Nadja Zöller
- Department of Dermatology, Venereology and Allergology, Johann Wolfgang Goethe-University, Theodor-Stern-Kai 7, D-60590 Frankfurt am Main, Germany
| | - Markus Meissner
- Department of Dermatology, Venereology and Allergology, Johann Wolfgang Goethe-University, Theodor-Stern-Kai 7, D-60590 Frankfurt am Main, Germany
| |
Collapse
|
46
|
Liu L, Han X, Huang Q, Zhu X, Yang J, Liu H. Increased neuronal seizure activity correlates with excessive systemic inflammation in a rat model of severe preeclampsia. Hypertens Res 2016; 39:701-708. [DOI: 10.1038/hr.2016.53] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2015] [Revised: 03/22/2016] [Accepted: 03/29/2016] [Indexed: 12/15/2022]
|
47
|
Lee PY, Yang CH, Kao MC, Su NY, Tsai PS, Huang CJ. Phosphoinositide 3-kinase β, phosphoinositide 3-kinase δ, and phosphoinositide 3-kinase γ mediate the anti-inflammatory effects of magnesium sulfate. J Surg Res 2015; 197:390-7. [DOI: 10.1016/j.jss.2015.04.051] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2014] [Revised: 03/17/2015] [Accepted: 04/14/2015] [Indexed: 01/08/2023]
|
48
|
A multicenter randomized controlled trial of intravenous magnesium for sickle cell pain crisis in children. Blood 2015; 126:1651-7. [PMID: 26232172 DOI: 10.1182/blood-2015-05-647107] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2015] [Accepted: 07/15/2015] [Indexed: 11/20/2022] Open
Abstract
Magnesium, a vasodilator, anti-inflammatory, and pain reliever, could alter the pathophysiology of sickle cell pain crises. We hypothesized that intravenous magnesium would shorten length of stay, decrease opioid use, and improve health-related quality of life (HRQL) for pediatric patients hospitalized with sickle cell pain crises. The Magnesium for Children in Crisis (MAGiC) study was a randomized, double-blind, placebo-controlled trial of intravenous magnesium vs normal saline placebo conducted at 8 sites within the Pediatric Emergency Care Applied Research Network (PECARN). Children 4 to 21 years old with hemoglobin SS or Sβ(0) thalassemia requiring hospitalization for pain were eligible. Children received 40 mg/kg of magnesium or placebo every 8 hours for up to 6 doses plus standard therapy. The primary outcome was length of stay in hours from the time of first study drug infusion, compared using a Van Elteren test. Secondary outcomes included opioid use and HRQL. Of 208 children enrolled, 204 received the study drug (101 magnesium, 103 placebo). Between-group demographics and prerandomization treatment were similar. The median interquartile range (IQR) length of stay was 56.0 (27.0-109.0) hours for magnesium vs 47.0 (24.0-99.0) hours for placebo (P = .24). Magnesium patients received 1.46 mg/kg morphine equivalents vs 1.28 mg/kg for placebo (P = .12). Changes in HRQL before discharge and 1 week after discharge were similar (P > .05 for all comparisons). The addition of intravenous magnesium did not shorten length of stay, reduce opioid use, or improve quality of life in children hospitalized for sickle cell pain crisis. This trial was registered at www.clinicaltrials.gov as #NCT01197417.
Collapse
|
49
|
Kamyar M, Manuck TA, Stoddard GJ, Varner MW, Clark EAS. Magnesium sulfate, chorioamnionitis, and neurodevelopment after preterm birth. BJOG 2015; 123:1161-6. [DOI: 10.1111/1471-0528.13460] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/02/2015] [Indexed: 11/28/2022]
Affiliation(s)
- M Kamyar
- Division of Maternal–Fetal Medicine Department of Obstetrics and Gynecology University of Utah Salt Lake City UT USA
| | - TA Manuck
- Division of Maternal–Fetal Medicine Department of Obstetrics and Gynecology University of Utah Salt Lake City UT USA
| | - GJ Stoddard
- Division of Epidemiology Department of Internal Medicine University of Utah School of Medicine Salt Lake City UT USA
| | - MW Varner
- Division of Maternal–Fetal Medicine Department of Obstetrics and Gynecology University of Utah Salt Lake City UT USA
| | - EAS Clark
- Division of Maternal–Fetal Medicine Department of Obstetrics and Gynecology University of Utah Salt Lake City UT USA
| |
Collapse
|
50
|
Da Silva MS, Rudkowska I. Dairy nutrients and their effect on inflammatory profile in molecular studies. Mol Nutr Food Res 2015; 59:1249-63. [DOI: 10.1002/mnfr.201400569] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2014] [Revised: 12/23/2014] [Accepted: 12/29/2014] [Indexed: 12/17/2022]
Affiliation(s)
- Marine S. Da Silva
- Department of Endocrinology and Nephrology; CHU de Québec Research Center; Quebec QC Canada
| | - Iwona Rudkowska
- Department of Endocrinology and Nephrology; CHU de Québec Research Center; Quebec QC Canada
| |
Collapse
|