1
|
Zhao Z, Xu W, Teng G, Xu X, Lu B, Zhou H, Wang L, Liu Y, Xu S, Wang Q, Ma W. Blood detection of autoimmune encephalitis based on laser-induced breakdown spectroscopy and Raman spectroscopy. Anal Chim Acta 2025; 1353:343948. [PMID: 40221195 DOI: 10.1016/j.aca.2025.343948] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2024] [Revised: 03/05/2025] [Accepted: 03/16/2025] [Indexed: 04/14/2025]
Abstract
BACKGROUND Recently, the incidence range of autoimmune encephalitis (AE) in people has rapidly expanded, and the diagnosis procedure of clinical criteria for AE remains complicated. Herein, with advantages of rapid speed, simple pre-treatment, and slightly destructive or non-destructive analysis, the feasibility of integrating laser-induced breakdown spectroscopy (LIBS) and Raman techniques to identify blood of AE patients was explored, and the mechanisms of medical diagnosis from atomic and molecular perspectives were further analyzed. RESULTS In the experiment, etched mesh silicon wafers were used as serum substrates to reduce the spectral variability during measurements. Totally, 1785 LIBS spectra and 1785 Raman spectra were collected from 119 people (79 healthy people and 40 AE patients), respectively. Fusion spectra were formed by connecting LIBS spectra in series behind with Raman spectra. With mutual information (MI) method, 537 features were selected from fusion spectra, and the accuracy and test time of long short-term memory model using these features were 95.04 % and 0.95 s, an improvement by 14.36 %, 8.03 %, 2.22 % and 0.48 s, 0.08 s, 0.55 s compared to using LIBS spectra, Raman spectra and fusion spectra, respectively. Besides, the correlations between spectra and cytokines were analyzed by the Pearson's correlation coefficient. Both metal atoms such as Na and K and molecules such as tryptophan, deoxyribose and phenylalanine were related to cytokines, corresponding to their MI importance in the AE diagnosis. SIGNIFICANCE We made the first attempt to identify AE blood using fusion of spectral techniques and analyze correlation mechanism between spectra and cytokines. All results demonstrated that it is feasible to accurately identify AE serum by fusing LIBS and Raman techniques, which is expected to effectively assist the clinical diagnosis of AE in the future.
Collapse
Affiliation(s)
- Zhifang Zhao
- School of Optics and Photonics, Beijing Institute of Technology, Beijing, 100081, China; Key Laboratory of Photonic Information Technology, Ministry of Industry and Information Technology, Beijing Institute of Technology, Beijing, 100081, China; National Key Laboratory on Near-surface Detection, Beijing, 10072, China
| | - Wangshu Xu
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, 100160, China
| | - Geer Teng
- School of Optics and Photonics, Beijing Institute of Technology, Beijing, 100081, China; Key Laboratory of Photonic Information Technology, Ministry of Industry and Information Technology, Beijing Institute of Technology, Beijing, 100081, China; National Key Laboratory on Near-surface Detection, Beijing, 10072, China; Institute of Biomedical Engineering, Department of Engineering Science, University of Oxford, Oxford, OX3 7LD, United Kingdom
| | - Xiangjun Xu
- School of Optics and Photonics, Beijing Institute of Technology, Beijing, 100081, China; Key Laboratory of Photonic Information Technology, Ministry of Industry and Information Technology, Beijing Institute of Technology, Beijing, 100081, China; National Key Laboratory on Near-surface Detection, Beijing, 10072, China
| | - Bingheng Lu
- School of Optics and Photonics, Beijing Institute of Technology, Beijing, 100081, China; Key Laboratory of Photonic Information Technology, Ministry of Industry and Information Technology, Beijing Institute of Technology, Beijing, 100081, China; National Key Laboratory on Near-surface Detection, Beijing, 10072, China
| | - Hao Zhou
- School of Optics and Photonics, Beijing Institute of Technology, Beijing, 100081, China; Key Laboratory of Photonic Information Technology, Ministry of Industry and Information Technology, Beijing Institute of Technology, Beijing, 100081, China; National Key Laboratory on Near-surface Detection, Beijing, 10072, China
| | - Leifu Wang
- School of Optics and Photonics, Beijing Institute of Technology, Beijing, 100081, China; Key Laboratory of Photonic Information Technology, Ministry of Industry and Information Technology, Beijing Institute of Technology, Beijing, 100081, China; National Key Laboratory on Near-surface Detection, Beijing, 10072, China
| | - Yuge Liu
- School of Optics and Photonics, Beijing Institute of Technology, Beijing, 100081, China; Key Laboratory of Photonic Information Technology, Ministry of Industry and Information Technology, Beijing Institute of Technology, Beijing, 100081, China; National Key Laboratory on Near-surface Detection, Beijing, 10072, China
| | - Shuai Xu
- School of Optics and Photonics, Beijing Institute of Technology, Beijing, 100081, China; Key Laboratory of Photonic Information Technology, Ministry of Industry and Information Technology, Beijing Institute of Technology, Beijing, 100081, China; National Key Laboratory on Near-surface Detection, Beijing, 10072, China
| | - Qianqian Wang
- School of Optics and Photonics, Beijing Institute of Technology, Beijing, 100081, China; Key Laboratory of Photonic Information Technology, Ministry of Industry and Information Technology, Beijing Institute of Technology, Beijing, 100081, China; National Key Laboratory on Near-surface Detection, Beijing, 10072, China.
| | - Wenping Ma
- Department of Neurosurgery, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, 100045, Beijing, China.
| |
Collapse
|
2
|
Zhang J, Wang X, Fang J, Li Y, Yu Y, Wang J, Sun B. Contributions of Dietary Patterns and Factors to Regulation of Rheumatoid Disease. Int J Mol Sci 2025; 26:2674. [PMID: 40141316 PMCID: PMC11942231 DOI: 10.3390/ijms26062674] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2024] [Revised: 03/04/2025] [Accepted: 03/13/2025] [Indexed: 03/28/2025] Open
Abstract
Rheumatoid arthritis (RA) is a systemic autoimmune disease that commonly causes pain in joints and the progressive destruction of cartilage and bone, which significantly reduces the quality of life and increases the social burden. However, there is still no cure for RA, so it is highly important to explore additional adjuvant treatment methods. Studies have indicated that malnutrition, changes in intestinal microbiota, and changes in immune status caused by dietary imbalance are directly related to the onset of RA, indicating that dietary intervention may offer a simple, economical, and practical avenue to relieve RA. Therefore, in this review, we discuss the pathogenesis of RA and summarize the influence of different dietary patterns on RA. In particular, we pointed out that high-fat, high-sugar, and high-salt diets contribute to RA progression, whereas the Mediterranean diet (MD) is beneficial for preventing RA. Furthermore, the ingredients of food, such as dietary fiber, probiotics, and vitamins, help reduce the level of inflammation and relieve joint pain, which may play critical roles in the treatment of RA. Therefore, dietary intervention provides a potential effective approach for adjuvant therapy of RA.
Collapse
Affiliation(s)
- Jingjie Zhang
- Key Laboratory of Geriatric Nutrition and Health, Ministry of Education, Beijing Technology and Business University, Beijing 100048, China; (J.Z.); (X.W.); (J.F.); (Y.L.); (B.S.)
- China-Canada Joint Laboratory of Food Nutrition and Health, Beijing Technology and Business University, Beijing 100048, China
- Key Laboratory of Special Food Supervision Technology for State Market Regulation, Beijing Technology and Business University, Beijing 100048, China
| | - Xueli Wang
- Key Laboratory of Geriatric Nutrition and Health, Ministry of Education, Beijing Technology and Business University, Beijing 100048, China; (J.Z.); (X.W.); (J.F.); (Y.L.); (B.S.)
- China-Canada Joint Laboratory of Food Nutrition and Health, Beijing Technology and Business University, Beijing 100048, China
- Key Laboratory of Special Food Supervision Technology for State Market Regulation, Beijing Technology and Business University, Beijing 100048, China
| | - Juan Fang
- Key Laboratory of Geriatric Nutrition and Health, Ministry of Education, Beijing Technology and Business University, Beijing 100048, China; (J.Z.); (X.W.); (J.F.); (Y.L.); (B.S.)
- China-Canada Joint Laboratory of Food Nutrition and Health, Beijing Technology and Business University, Beijing 100048, China
- Key Laboratory of Special Food Supervision Technology for State Market Regulation, Beijing Technology and Business University, Beijing 100048, China
| | - Yingying Li
- Key Laboratory of Geriatric Nutrition and Health, Ministry of Education, Beijing Technology and Business University, Beijing 100048, China; (J.Z.); (X.W.); (J.F.); (Y.L.); (B.S.)
- China-Canada Joint Laboratory of Food Nutrition and Health, Beijing Technology and Business University, Beijing 100048, China
- Key Laboratory of Special Food Supervision Technology for State Market Regulation, Beijing Technology and Business University, Beijing 100048, China
| | - Yonghui Yu
- Key Laboratory of Geriatric Nutrition and Health, Ministry of Education, Beijing Technology and Business University, Beijing 100048, China; (J.Z.); (X.W.); (J.F.); (Y.L.); (B.S.)
- China-Canada Joint Laboratory of Food Nutrition and Health, Beijing Technology and Business University, Beijing 100048, China
- Key Laboratory of Special Food Supervision Technology for State Market Regulation, Beijing Technology and Business University, Beijing 100048, China
| | - Jing Wang
- Key Laboratory of Geriatric Nutrition and Health, Ministry of Education, Beijing Technology and Business University, Beijing 100048, China; (J.Z.); (X.W.); (J.F.); (Y.L.); (B.S.)
- China-Canada Joint Laboratory of Food Nutrition and Health, Beijing Technology and Business University, Beijing 100048, China
- Key Laboratory of Special Food Supervision Technology for State Market Regulation, Beijing Technology and Business University, Beijing 100048, China
| | - Baoguo Sun
- Key Laboratory of Geriatric Nutrition and Health, Ministry of Education, Beijing Technology and Business University, Beijing 100048, China; (J.Z.); (X.W.); (J.F.); (Y.L.); (B.S.)
- China-Canada Joint Laboratory of Food Nutrition and Health, Beijing Technology and Business University, Beijing 100048, China
- Key Laboratory of Special Food Supervision Technology for State Market Regulation, Beijing Technology and Business University, Beijing 100048, China
| |
Collapse
|
3
|
Masenga SK, Wandira N, Cattivelli-Murdoch G, Saleem M, Beasley H, Hinton A, Ertuglu LA, Mwesigwa N, Kleyman TR, Kirabo A. Salt sensitivity of blood pressure: mechanisms and sex-specific differences. Nat Rev Cardiol 2025:10.1038/s41569-025-01135-0. [PMID: 39984695 DOI: 10.1038/s41569-025-01135-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 01/31/2025] [Indexed: 02/23/2025]
Abstract
Salt sensitivity of blood pressure (SSBP) is an independent risk factor for cardiovascular disease in individuals with or without hypertension. However, the mechanisms and management of SSBP remain unclear, mainly because the diagnosis of this condition relies on salt loading-depletion protocols that are not feasible in the clinic. The prevalence of hypertension is lower in premenopausal women than in men, but this sex-specific difference is reversed after menopause. Whether excessive SSBP in women at any age contributes to this reversal is unknown, but many clinical studies that have rigorously assessed for SSBP using salt loading-depletion protocols have confirmed that SSBP is more prevalent in women than in men, including during premenopausal age. In this Review, we discuss sex-specific mechanisms of SSBP. We describe sex-related differences in renal transporters, hypertensive pregnancy, SSBP in autoimmune disorders and mitogen-activated protein kinase signalling pathways, and highlight limitations and lessons learned from Dahl salt-sensitive rat models.
Collapse
Affiliation(s)
- Sepiso K Masenga
- HAND research Group, Department of Pathology and Physiological Sciences, School of Medicine and Health Sciences, Mulungushi University, Livingstone, Zambia.
- Vanderbilt Institute for Global Health, Vanderbilt University Medical Center, Nashville, TN, USA.
| | - Nelson Wandira
- Vanderbilt Mater of Public Health Program, Vanderbilt University Medical Center, Nashville, TN, USA
| | | | - Mohammad Saleem
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Heather Beasley
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, USA
| | - Antentor Hinton
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, USA
| | - Lale A Ertuglu
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Naome Mwesigwa
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Thomas R Kleyman
- Renal-Electrolyte Division, Department of Medicine, Department of Cell Biology, and Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Annet Kirabo
- Vanderbilt Institute for Global Health, Vanderbilt University Medical Center, Nashville, TN, USA.
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA.
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, USA.
- Vanderbilt Center for Immunobiology, Vanderbilt University Medical Center, Nashville, TN, USA.
- Vanderbilt Institute for Infection, Immunology and Inflammation, Vanderbilt University Medical Center, Nashville, TN, USA.
| |
Collapse
|
4
|
An P, Wang S, Liu L, Li X, Lv X. The association between dietary sodium density and Hashimoto's thyroiditis in US adults. Front Nutr 2025; 12:1508195. [PMID: 39911805 PMCID: PMC11794099 DOI: 10.3389/fnut.2025.1508195] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Accepted: 01/09/2025] [Indexed: 02/07/2025] Open
Abstract
Background Hashimoto's thyroiditis (HT) is an autoimmune thyroid disease characterized by the presence of antibodies against thyroid-specific antigens. Dietary sodium intake has been implicated in the development of several autoimmune diseases, but its association with HT remains unclear. Methods This study investigates the relationship between dietary sodium density (the ratio of sodium to energy intake) and HT using data from the National Health and Nutrition Examination Survey (NHANES) from 2007 to 2012. A cross-sectional study was conducted using NHANES data, focusing on adults aged 20 years and older with available dietary and thyroid profile data. Sodium density was calculated from two 24-h dietary recall interviews. Logistic regression models were used to evaluate the associations of sodium density with HT, thyroid peroxidase antibody (TPOAb) and thyroglobulin antibody (TgAb). Restricted cubic spline (RCS) analyses were performed to explore non-linear relationships. Results A total of 6,258 participants were included, with 576 (9.20%) diagnosed with HT. An additional unit of dietary sodium density was associated with a 24% increased risk of HT (OR 1.24, 95% CI 1.01-1.50) in adjusted model. A breakpoint at 2.43 mg/kcal in dietary sodium density was identified using a piecewise regression model. Below this threshold, HT risk increased with rising sodium density, while above it, the risk plateaued. Higher sodium density was also associated with increased TPOAb positivity (OR 1.28, 95% CI 1.05-1.56), but not TgAb positivity. Conclusion Elevated dietary sodium density is associated with an increased risk of HT and TPOAb positivity, suggesting that sodium intake may play a role in the pathogenesis of HT.
Collapse
Affiliation(s)
- Peilin An
- Department of Health Management Center, Chongqing General Hospital, Chongqing University, Chongqing, China
| | - Silin Wang
- Department of Electrical Engineering, Chalmers University of Technology, Gothenburg, Sweden
| | - Lingyun Liu
- Department of Health Management Center, Chongqing General Hospital, Chongqing University, Chongqing, China
| | - Xuelin Li
- Department of Health Management Center, Chongqing General Hospital, Chongqing University, Chongqing, China
| | - Xin Lv
- Department of Clinical Nutrition, The Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, China
- Yangzhou Key Laboratory of Preventive and Translational Medicine for Geriatric Disease (Frailty), The Affiliated Hospital of Yangzhou University, Yangzhou, China
| |
Collapse
|
5
|
Mutchler AL, Haynes AP, Saleem M, Jamison S, Khan MM, Ertuglu L, Kirabo A. Epigenetic Regulation of Innate and Adaptive Immune Cells in Salt-Sensitive Hypertension. Circ Res 2025; 136:232-254. [PMID: 39819017 PMCID: PMC11750173 DOI: 10.1161/circresaha.124.325439] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/19/2025]
Abstract
Access to excess dietary sodium has heightened the risk of cardiovascular diseases, particularly affecting individuals with salt sensitivity of blood pressure. Our research indicates that innate antigen-presenting immune cells contribute to rapid blood pressure increases in response to excess sodium intake. Emerging evidence suggests that epigenetic reprogramming, with subsequent transcriptional and metabolic changes, of innate immune cells allows these cells to have a sustained response to repetitive stimuli. Epigenetic mechanisms also steer T-cell differentiation in response to innate immune signaling. Immune cells respond to environmental and nutritional cues, such as salt, promoting epigenetic regulation changes. This article aims to identify and discuss the role of epigenetic mechanisms in the immune system contributing to salt-sensitive hypertension.
Collapse
Affiliation(s)
- Ashley L. Mutchler
- Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Alexandria Porcia Haynes
- Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Mohammad Saleem
- Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | | | - Mohd Mabood Khan
- Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Lale Ertuglu
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Annet Kirabo
- Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
- Department of Molecular Physiology & Biophysics, Vanderbilt University, Nashville, TN 37212-8802, USA
- Vanderbilt Center for Immunobiology
- Vanderbilt Institute for Infection, Immunology and Inflammation
- Vanderbilt Institute for Global Health
| |
Collapse
|
6
|
Jiang X, Yang Y, Ho CT, Muhoza B, Liu Q, Song S. Exploring the salty peptides of enzymatically hydrolyzed Volvariella volvacea protein: Structural analysis and taste perception insights. Food Res Int 2024; 197:115155. [PMID: 39593367 DOI: 10.1016/j.foodres.2024.115155] [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: 08/06/2024] [Revised: 09/21/2024] [Accepted: 09/26/2024] [Indexed: 11/28/2024]
Abstract
This study aimed to prepare and screen salty peptides and elucidate the factors triggering their saltiness properties and saltiness-enhancing effects. Electronic tongue results indicated that samples treated with Alcalase (AJ) exhibited the highest saltiness intensity (5.05 ± 0.11) and greater saltiness-enhancing compared to those treated with Flavourzyme (AF), Neutrase (AZ), and Papain (AM). Peptides identified by LC-MS/MS were docked to the TMC4 receptor, resulting in 23 peptides with good docked results. The key amino acid residues (Thr431, Arg433, Phe440, Phe432, and Ser273) were discovered. The salty peptides WPGFK (633.75 Da), YFDWPGFK (1059.17 Da), and YFDWPGFKT (1160.27 Da) were selected for salt reduction and molecular characterization studies. Electronic tongue results showed that the 0.01 % WPGFK solution matched the saltiness of the 0.24 % NaCl solution and demonstrated superior saltiness-enhancing compared to YFDWPGFK and YFDWPGFKT. Primary sequence analysis of three salty peptides suggested that the WPG amino acid segment might play a crucial role in generating saltiness. Circular dichroism (CD) analysis of the secondary structure indicated that increased random coil and decreased β-turn contents resulted in higher saltiness perception and increased random coil content led to greater saltiness-enhancing ability.
Collapse
Affiliation(s)
- Xiaoyu Jiang
- School of Perfume and Aroma Technology, Shanghai Institute of Technology, 100 Haiquan Road, Shanghai 201418, China
| | - Yang Yang
- School of Perfume and Aroma Technology, Shanghai Institute of Technology, 100 Haiquan Road, Shanghai 201418, China
| | - Chi-Tang Ho
- Department of Food Science, Rutgers University, 65 Dudley Road, New Brunswick, NJ 08901, USA
| | - Bertrand Muhoza
- College of Food Science, Northeast Agricultural University, 600 Changjiang Road, Harbin 150030, Heilongjiang, China
| | - Qian Liu
- School of Perfume and Aroma Technology, Shanghai Institute of Technology, 100 Haiquan Road, Shanghai 201418, China
| | - Shiqing Song
- School of Perfume and Aroma Technology, Shanghai Institute of Technology, 100 Haiquan Road, Shanghai 201418, China.
| |
Collapse
|
7
|
Hu Z, Tian Y, Yang A, Song X. High sodium intake increases interstitial lung disease and pulmonary sarcoidosis based on the Global Burden of Disease study 1999-2019. Sci Rep 2024; 14:25891. [PMID: 39472704 PMCID: PMC11522497 DOI: 10.1038/s41598-024-77769-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Accepted: 10/25/2024] [Indexed: 11/02/2024] Open
Abstract
This study investigated the relationships between dietary sodium intake and the incidence and prevalence of interstitial lung disease (ILD) and pulmonary sarcoidosis using data from the Global Burden of Diseases, Injuries, and Risk Factors Study 2019. This study assessed the strength of the abovementioned relationships via LASSO analysis and a generalized additive model with Poisson regression and determined the nonlinear and lagged effects via a distributed lag nonlinear model (DLNM). In the past three decades, global dietary sodium intake has decreased gradually. Two LASSO and generalized additive analyses both suggested that dietary sodium intake is obviously correlated with the incidence and prevalence of ILD and pulmonary sarcoidosis. The overall exposure‒response curve revealed a dose‒effect relationship between dietary sodium intake and the incidence and prevalence of ILD and pulmonary sarcoidosis. The maximum lag-specific RR of extremely high dietary sodium intake was 1.75 (95% CI: 1.61-1.91, lag 0 year) for incidence and 3.19 (95% CI: 2.24-4.53, lag 0 year) for prevalence relative to the reference. Our study suggests that dietary sodium intake is positively associated with the incidence and prevalence of ILD and pulmonary sarcoidosis. These findings may have important policy implications for dietary sodium intake-reduction strategies to decrease the burden of respiratory diseases and promote public health.
Collapse
Affiliation(s)
- Zhigang Hu
- Department of Respiratory and Critical Care Medicine, The First College of Clinical Medicine Science, China Three Gorges University, Yichang, 443003, People's Republic of China.
- Department of Respiratory and Critical Care Medicine, Yichang Central People's Hospital, Yichang, 443003, People's Republic of China.
- Clinical Medical Research Center for Precision Diagnosis and Treatment of Lung Cancer and Management of Advance Cancer Pain of Hubei Province, Zhijiang, People's Republic of China.
- Department of Respiratory and Critical Care Medicine, Yichang Central People's Hospital at Zhijiang, Zhijiang, 443200, People's Republic of China.
| | - Yufeng Tian
- Department of Teaching Office, Three Gorges University, Yichang, 443003, People's Republic of China
| | - Ailan Yang
- Department of Respiratory and Critical Care Medicine, Yichang Central People's Hospital at Zhijiang, Zhijiang, 443200, People's Republic of China
| | - Xinyu Song
- Department of Respiratory and Critical Care Medicine, The First College of Clinical Medicine Science, China Three Gorges University, Yichang, 443003, People's Republic of China.
- Department of Respiratory and Critical Care Medicine, Yichang Central People's Hospital, Yichang, 443003, People's Republic of China.
- Clinical Medical Research Center for Precision Diagnosis and Treatment of Lung Cancer and Management of Advance Cancer Pain of Hubei Province, Zhijiang, People's Republic of China.
| |
Collapse
|
8
|
Chernova I. Lupus Nephritis: Immune Cells and the Kidney Microenvironment. KIDNEY360 2024; 5:1394-1401. [PMID: 39120952 PMCID: PMC11441818 DOI: 10.34067/kid.0000000000000531] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Accepted: 07/29/2024] [Indexed: 08/11/2024]
Abstract
Lupus nephritis (LN) is the most common major organ manifestation of the autoimmune disease SLE (lupus), with 10% of those afflicted progressing to ESKD. The kidney in LN is characterized by a significant immune infiltrate and proinflammatory cytokine milieu that affects intrinsic renal cells and is, in part, responsible for the tissue damage observed in LN. It is now increasingly appreciated that LN is not due to unidirectional immune cell activation with subsequent kidney damage. Rather, the kidney microenvironment influences the recruitment, survival, differentiation, and activation of immune cells, which, in turn, modify kidney cell function. This review covers how the biochemical environment of the kidney ( i.e ., low oxygen tension and hypertonicity) and unique kidney cell types affect the intrarenal immune cells in LN. The pathways used by intrinsic renal cells to interact with immune cells, such as antigen presentation and cytokine production, are discussed in detail. An understanding of these mechanisms can lead to the design of more kidney-targeted treatments and the avoidance of systemic immunosuppressive effects and may represent the next frontier of LN therapies.
Collapse
Affiliation(s)
- Irene Chernova
- Section of Nephrology, Department of Internal Medicine, Yale School of Medicine, New Haven, Connecticut
| |
Collapse
|
9
|
Mo Q, Bolideei M, Rong SJ, Luo JH, Yang CL, Lu WY, Chen QJ, Zhao JW, Wang FX, Wang T, Li Y, Luo X, Zhang S, Xiong F, Yu QL, Zhang ZY, Liu SW, Sun F, Dong LL, Wang CY. GSK2334470 attenuates high salt-exacerbated rheumatoid arthritis progression by restoring Th17/Treg homeostasis. iScience 2024; 27:109798. [PMID: 38947509 PMCID: PMC11214488 DOI: 10.1016/j.isci.2024.109798] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Revised: 02/09/2024] [Accepted: 04/18/2024] [Indexed: 07/02/2024] Open
Abstract
High salt (HS) consumption is a risk factor for multiple autoimmune disorders via disturbing immune homeostasis. Nevertheless, the exact mechanisms by which HS exacerbates rheumatoid arthritis (RA) pathogenesis remain poorly defined. Herein, we found that heightened phosphorylation of PDPK1 and SGK1 upon HS exposure attenuated FoxO1 expression to enhance the glycolytic capacity of CD4 T cells, resulting in strengthened Th17 but compromised Treg program. GSK2334470 (GSK), a dual PDPK1/SGK1 inhibitor, effectively mitigated the HS-induced enhancement in glycolytic capacity and the overproduction of IL-17A. Therefore, administration of GSK markedly alleviated HS-exacerbated RA progression in collagen-induced arthritis (CIA) model. Collectively, our data indicate that HS consumption subverts Th17/Treg homeostasis through the PDPK1-SGK1-FoxO1 signaling, while GSK could be a viable drug against RA progression in clinical settings.
Collapse
Affiliation(s)
- Qian Mo
- Department of Respiratory and Critical Care Medicine, the Center for Biomedical Research, NHC Key Laboratory of Respiratory Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Sciences and Technology, Wuhan, China
- Department of Rheumatology, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, China
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan, China
| | - Mansoor Bolideei
- Department of Respiratory and Critical Care Medicine, the Center for Biomedical Research, NHC Key Laboratory of Respiratory Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Sciences and Technology, Wuhan, China
| | - Shan-Jie Rong
- Department of Respiratory and Critical Care Medicine, the Center for Biomedical Research, NHC Key Laboratory of Respiratory Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Sciences and Technology, Wuhan, China
| | - Jia-Hui Luo
- Department of Respiratory and Critical Care Medicine, the Center for Biomedical Research, NHC Key Laboratory of Respiratory Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Sciences and Technology, Wuhan, China
| | - Chun-Liang Yang
- Department of Respiratory and Critical Care Medicine, the Center for Biomedical Research, NHC Key Laboratory of Respiratory Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Sciences and Technology, Wuhan, China
| | - Wan-Ying Lu
- Department of Respiratory and Critical Care Medicine, the Center for Biomedical Research, NHC Key Laboratory of Respiratory Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Sciences and Technology, Wuhan, China
| | - Qi-Jie Chen
- Department of Respiratory and Critical Care Medicine, the Center for Biomedical Research, NHC Key Laboratory of Respiratory Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Sciences and Technology, Wuhan, China
| | - Jia-Wei Zhao
- Department of Respiratory and Critical Care Medicine, the Center for Biomedical Research, NHC Key Laboratory of Respiratory Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Sciences and Technology, Wuhan, China
| | - Fa-Xi Wang
- Department of Respiratory and Critical Care Medicine, the Center for Biomedical Research, NHC Key Laboratory of Respiratory Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Sciences and Technology, Wuhan, China
| | - Ting Wang
- Department of Respiratory and Critical Care Medicine, the Center for Biomedical Research, NHC Key Laboratory of Respiratory Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Sciences and Technology, Wuhan, China
| | - Yang Li
- Department of Respiratory and Critical Care Medicine, the Center for Biomedical Research, NHC Key Laboratory of Respiratory Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Sciences and Technology, Wuhan, China
| | - Xi Luo
- Department of Respiratory and Critical Care Medicine, the Center for Biomedical Research, NHC Key Laboratory of Respiratory Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Sciences and Technology, Wuhan, China
| | - Shu Zhang
- Department of Respiratory and Critical Care Medicine, the Center for Biomedical Research, NHC Key Laboratory of Respiratory Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Sciences and Technology, Wuhan, China
| | - Fei Xiong
- Department of Respiratory and Critical Care Medicine, the Center for Biomedical Research, NHC Key Laboratory of Respiratory Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Sciences and Technology, Wuhan, China
| | - Qi-Lin Yu
- Department of Respiratory and Critical Care Medicine, the Center for Biomedical Research, NHC Key Laboratory of Respiratory Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Sciences and Technology, Wuhan, China
| | - Zi-Yun Zhang
- Department of Rheumatology, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Shi-Wei Liu
- Shanxi Bethune Hospital, Shanxi Academy of Medical Science, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, the Key Laboratory of Endocrine and Metabolic Diseases of Shanxi Province, Taiyuan, China
| | - Fei Sun
- Department of Respiratory and Critical Care Medicine, the Center for Biomedical Research, NHC Key Laboratory of Respiratory Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Sciences and Technology, Wuhan, China
| | - Ling-Li Dong
- Department of Rheumatology, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Cong-Yi Wang
- Department of Respiratory and Critical Care Medicine, the Center for Biomedical Research, NHC Key Laboratory of Respiratory Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Sciences and Technology, Wuhan, China
- Shanxi Bethune Hospital, Shanxi Academy of Medical Science, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, the Key Laboratory of Endocrine and Metabolic Diseases of Shanxi Province, Taiyuan, China
| |
Collapse
|
10
|
Derluyn N, Foucart V, Verce M, Abdo R, Vaudoisey L, Lipski D, Flamand V, Everard A, Bruyns C, Willermain F. High salt diet alleviates disease severity in native experimental autoimmune uveitis. FRONTIERS IN OPHTHALMOLOGY 2024; 4:1370374. [PMID: 38984146 PMCID: PMC11182228 DOI: 10.3389/fopht.2024.1370374] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/14/2024] [Accepted: 04/19/2024] [Indexed: 07/11/2024]
Abstract
Background Recent studies reported a link between high salt diet (HSD) and clinical exacerbation in mouse models of autoimmune diseases, mainly through the induction of pathogenic Th17 cells and/or HSD-induced dysbiosis. However, the topic remains controversial and not fully understood. Purpose In this study, we investigated the effects of HSD on the development of experimental autoimmune uveitis (EAU) in C57BL/6J mice. Methods and results Unexpectedly, our data showed a significant attenuating effect of HSD on disease severity of native EAU, induced by direct immunization with IRBP peptide. That said, HSD had no effect on EAU disease severity induced by adoptive transfer of semi-purified auto-reactive IRBP-specific T lymphocytes. Accordingly, HSD did not affect IRBP-specific systemic afferent immune response as attested by no HSD-linked changes in T lymphocytes proliferation, cytokine production and Treg proportion. Gut microbiota analysis from cecal samples in naïve and EAU mice demonstrated that HSD affected differentially α-diversity between groups, whereas β-diversity was significantly modified in all groups. Unknown Tannerellaceae was the only taxon associated to HSD exposure in all treatment groups. Interestingly, a significantly higher abundance of unknown Gastranaerophilales, with potential anti-inflammatory properties, appeared in HSD-fed native EAU mice, only. Discussion In conclusion, our study suggests a possible impact of HSD on gut microbiota composition and consequently on development and clinical severity of EAU. Further studies are required to investigate the potential beneficial role of Gastranaerophilales in EAU.
Collapse
Affiliation(s)
- Naomi Derluyn
- Institute of Interdisciplinary Research (IRIBHM), Université Libre de Bruxelles (ULB), Brussels, Belgium
- Department of Ophthalmology, CHU Saint-Pierre, Université Libre de Bruxelles (ULB), Brussels, Belgium
- Department of Ophthalmology, CHU Brugmann, Université Libre de Bruxelles (ULB), Brussels, Belgium
| | - Vincent Foucart
- Institute of Interdisciplinary Research (IRIBHM), Université Libre de Bruxelles (ULB), Brussels, Belgium
- Department of Ophthalmology, CHU Saint-Pierre, Université Libre de Bruxelles (ULB), Brussels, Belgium
| | - Marko Verce
- Metabolism and Nutrition Research Group, Louvain Drug Research Institute, UCLouvain, Université Catholique de Louvain, Brussels, Belgium
- WELBIO Department, WEL Research Institute, Walloon Excellence in Life Sciences and BIOtechnology (WELBIO), Wavre, Belgium
| | - Rami Abdo
- Institute of Interdisciplinary Research (IRIBHM), Université Libre de Bruxelles (ULB), Brussels, Belgium
- Department of Ophthalmology, Hôpital Universitaire de Bruxelles (HUB) - Hôpital Erasme, Université Libre de Bruxelles (ULB), Brussels, Belgium
| | - Louis Vaudoisey
- Institute of Interdisciplinary Research (IRIBHM), Université Libre de Bruxelles (ULB), Brussels, Belgium
- Department of Ophthalmology, CHU Saint-Pierre, Université Libre de Bruxelles (ULB), Brussels, Belgium
| | - Deborah Lipski
- Institute of Interdisciplinary Research (IRIBHM), Université Libre de Bruxelles (ULB), Brussels, Belgium
- Department of Ophthalmology, Hôpital Universitaire de Bruxelles (HUB) - Hôpital Erasme, Université Libre de Bruxelles (ULB), Brussels, Belgium
| | - Véronique Flamand
- Institute for Medical Immunology, Université Libre de Bruxelles (ULB), Charleroi, Belgium
| | - Amandine Everard
- Metabolism and Nutrition Research Group, Louvain Drug Research Institute, UCLouvain, Université Catholique de Louvain, Brussels, Belgium
- WELBIO Department, WEL Research Institute, Walloon Excellence in Life Sciences and BIOtechnology (WELBIO), Wavre, Belgium
| | - Catherine Bruyns
- Institute of Interdisciplinary Research (IRIBHM), Université Libre de Bruxelles (ULB), Brussels, Belgium
| | - François Willermain
- Institute of Interdisciplinary Research (IRIBHM), Université Libre de Bruxelles (ULB), Brussels, Belgium
- Department of Ophthalmology, CHU Saint-Pierre, Université Libre de Bruxelles (ULB), Brussels, Belgium
- Department of Ophthalmology, CHU Brugmann, Université Libre de Bruxelles (ULB), Brussels, Belgium
| |
Collapse
|
11
|
Sang J, Dhakal S, Shrestha B, Nath DK, Kim Y, Ganguly A, Montell C, Lee Y. A single pair of pharyngeal neurons functions as a commander to reject high salt in Drosophila melanogaster. eLife 2024; 12:RP93464. [PMID: 38573740 PMCID: PMC10994663 DOI: 10.7554/elife.93464] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/05/2024] Open
Abstract
Salt (NaCl), is an essential nutrient for survival, while excessive salt can be detrimental. In the fruit fly, Drosophila melanogaster, internal taste organs in the pharynx are critical gatekeepers impacting the decision to accept or reject a food. Currently, our understanding of the mechanism through which pharyngeal gustatory receptor neurons (GRNs) sense high salt are rudimentary. Here, we found that a member of the ionotropic receptor family, Ir60b, is expressed exclusively in a pair of GRNs activated by high salt. Using a two-way choice assay (DrosoX) to measure ingestion volume, we demonstrate that IR60b and two co-receptors IR25a and IR76b are required to prevent high salt consumption. Mutants lacking external taste organs but retaining the internal taste organs in the pharynx exhibit much higher salt avoidance than flies with all taste organs but missing the three IRs. Our findings highlight the vital role for IRs in a pharyngeal GRN to control ingestion of high salt.
Collapse
Affiliation(s)
- Jiun Sang
- Department of Bio and Fermentation Convergence Technology, Kookmin UniversitySeoulRepublic of Korea
| | - Subash Dhakal
- Department of Bio and Fermentation Convergence Technology, Kookmin UniversitySeoulRepublic of Korea
| | - Bhanu Shrestha
- Department of Bio and Fermentation Convergence Technology, Kookmin UniversitySeoulRepublic of Korea
| | - Dharmendra Kumar Nath
- Department of Bio and Fermentation Convergence Technology, Kookmin UniversitySeoulRepublic of Korea
| | - Yunjung Kim
- Department of Bio and Fermentation Convergence Technology, Kookmin UniversitySeoulRepublic of Korea
| | - Anindya Ganguly
- Neuroscience Research Institute and Department of Molecular, Cellular and Developmental Biology, University of California, Santa BarbaraSanta BarbaraUnited States
| | - Craig Montell
- Neuroscience Research Institute and Department of Molecular, Cellular and Developmental Biology, University of California, Santa BarbaraSanta BarbaraUnited States
| | - Youngseok Lee
- Department of Bio and Fermentation Convergence Technology, Kookmin UniversitySeoulRepublic of Korea
| |
Collapse
|
12
|
Sang J, Dhakal S, Shrestha B, Nath DK, Kim Y, Ganguly A, Montell C, Lee Y. A single pair of pharyngeal neurons functions as a commander to reject high salt in Drosophila melanogaster. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.10.17.562703. [PMID: 37904986 PMCID: PMC10614918 DOI: 10.1101/2023.10.17.562703] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/02/2023]
Abstract
Salt is an essential nutrient for survival, while excessive NaCl can be detrimental. In the fruit fly, Drosophila melanogaster, internal taste organs in the pharynx are critical gatekeepers impacting the decision to accept or reject a food. Currently, our understanding of the mechanism through which pharyngeal gustatory receptor neurons (GRNs) sense high salt are rudimentary. Here, we found that a member of the ionotropic receptor family, Ir60b, is expressed exclusively in a pair of GRNs activated by high salt. Using a two-way choice assay (DrosoX) to measure ingestion volume, we demonstrate that IR60b and two coreceptors IR25a and IR76b, are required to prevent high salt consumption. Mutants lacking external taste organs but retaining the internal taste organs in the pharynx exhibit much higher salt avoidance than flies with all taste organs but missing the three IRs. Our findings highlight the vital role for IRs in a pharyngeal GRN to control ingestion of high salt.
Collapse
Affiliation(s)
- Jiun Sang
- Department of Bio and Fermentation Convergence Technology, Kookmin University, Seoul, 02707, Republic of Korea
- These authors contributed equally
| | - Subash Dhakal
- Department of Bio and Fermentation Convergence Technology, Kookmin University, Seoul, 02707, Republic of Korea
- These authors contributed equally
| | - Bhanu Shrestha
- Department of Bio and Fermentation Convergence Technology, Kookmin University, Seoul, 02707, Republic of Korea
| | - Dharmendra Kumar Nath
- Department of Bio and Fermentation Convergence Technology, Kookmin University, Seoul, 02707, Republic of Korea
| | - Yunjung Kim
- Department of Bio and Fermentation Convergence Technology, Kookmin University, Seoul, 02707, Republic of Korea
| | - Anindya Ganguly
- Neuroscience Research Institute and Department of Molecular, Cellular and Developmental Biology, University of California, Santa Barbara, Santa Barbara, CA United States
| | - Craig Montell
- Neuroscience Research Institute and Department of Molecular, Cellular and Developmental Biology, University of California, Santa Barbara, Santa Barbara, CA United States
| | - Youngseok Lee
- Department of Bio and Fermentation Convergence Technology, Kookmin University, Seoul, 02707, Republic of Korea
- Lead Contract
| |
Collapse
|
13
|
Jula A. Sodium - a systematic review for Nordic Nutrition Recommendations 2023. Food Nutr Res 2024; 68:10319. [PMID: 38327996 PMCID: PMC10845896 DOI: 10.29219/fnr.v68.10319] [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: 02/26/2022] [Revised: 11/25/2022] [Accepted: 10/25/2023] [Indexed: 02/09/2024] Open
Abstract
Blood pressure (BP) rises along with increasing sodium intake from early childhood to late adulthood, and leads to hypertension among most men and women living in Nordic and Baltic countries. Elevated BP is the leading global risk factor for premature deaths and disability-adjusted life-years. A reduction in sodium intake is essential in the prevention of hypertension in individuals, in the lowering of BP levels, in the treatment of hypertensive individuals, and in decreasing risks associated with elevated BP. There is a progressive linear dose-response relationship between sodium intake and BP beginning from a sodium intake of less than 0.8 g/day. Sodium reduction decreases BP linearly by a dose-response manner down to a sodium intake level of less than 2 g/day. Randomised intervention studies with a duration of at least 4 weeks confirm the efficiency and safety of reducing blood sodium intake to a level of less than 2 g/day. Results from prospective cohort studies show that higher sodium intake is positively associated with an increased risk of stroke and cardiovascular events and mortality among the general adult population, and the associations are linear in studies using proper sodium assessment methods. Analyses assessing sodium intake using at least two 24-h urine samples have shown a linear positive relationship between sodium intake and the risk of a cardiovascular event or death. Based on an overall evaluation of the available data, a limitation of the sodium intake to 2.0 g/day is suggested for adults. The optimal sodium intake level would be probably about 1.5 g/day. Sodium intake recommended for children can be extrapolated from the recommended sodium intake for adults. According to national dietary surveys, the average sodium intakes in Nordic countries range in adult men from 3.6 to 4.4 g/day and in adult women from 2.6. to 3.2 g/day, and in Baltic countries in men from 2.6 to 5.1 g/day and in women from 1.8 to 3.6 g/day.
Collapse
Affiliation(s)
- Antti Jula
- Department of Clinical Medicine, University of Turku, Turku, Finland
| |
Collapse
|
14
|
Armas AJM, Aranda JAE, Arcos HS, Arellano FL, Arguelles CBL, Arreza AMM, Arriza MAG, Ascan CKA, Torres GCS. Assessing Dietary Salt Intake and Pilot-Testing a Home-Based Intervention to Lower Salt Intake Among Filipino College Students. J Community Health Nurs 2024; 41:57-72. [PMID: 37943282 DOI: 10.1080/07370016.2023.2277839] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2023]
Abstract
PURPOSE To assess the knowledge, attitude, and behaviors related to dietary salt intake and develop a home-based intervention that helps lower salt intake among Filipino college students. DESIGN Two-phase program development study utilizing descriptive and quasi-experimental design. METHODS Phase 1 determined the knowledge, attitude, and behaviors alongside the frequency of dietary salt intake of a consecutive sample (n = 118). Phase 2 involved the development and pilot-testing of a home-based intervention wherein a random sample (n = 35) selected from Phase 1 joined for pilot-testing. The study was conducted from October - November 2021 and utilized valid and reliable measures to test the knowledge, attitude, and behavior of the participants. FINDINGS Initial assessment showed knowledge regarding health risks of high salt intake and positive attitude toward health-promoting behaviors. Post-intervention showed improvement in attitude (p = 0.0004) and behavior (p = 0.001) related to dietary salt intake alongside health literacy (p = 0.036). There was no significant change in knowledge (p = 0.054). CONCLUSION The home-based intervention involving the use of health education materials is successful in improving dietary salt intake patterns. CLINICAL EVIDENCE Community health nurses may utilize home-based interventions to help develop health-promoting behaviors among young adults.
Collapse
|
15
|
Afsar B, Afsar RE. Salt Behind the Scenes of Systemic Lupus Erythematosus and Rheumatoid Arthritis. Curr Nutr Rep 2023; 12:830-844. [PMID: 37980312 DOI: 10.1007/s13668-023-00509-5] [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] [Accepted: 11/09/2023] [Indexed: 11/20/2023]
Abstract
PURPOSE OF REVIEW Sodium is vital for human health. High salt intake is a global health problem and is associated with cardiovascular morbidity and mortality. Recent evidence suggests that both innate and adaptive immune systems are affected by sodium. In general, excess salt intake drives immune cells toward a pro-inflammatory phenotype. The incidence of autoimmune diseases, including systemic lupus erythematosus (SLE) and rheumatoid arthritis (RA), is steadily increasing. As excess salt induces a pro-inflammatory state, increased salt intake may have impacts on autoimmune diseases. The relationship between salt intake and autoimmune diseases is most widely studied in patients with SLE or RA. This review aimed to summarize the relationship between salt intake and SLE and RA. RECENT FINDINGS Most, but not all, of these studies showed that high salt intake might promote SLE by M1 macrophage shift, increase in Th17/Treg cell ratio, activation of dendritic and follicular helper T cells, and increased secretion of pro-inflammatory cytokines. In RA, apart from driving immune cells toward a pro-inflammatory state, high salt intake also influences cellular signaling pathways, including receptor activator of nuclear factor κB ligand (RANKL), Rho GTPases, and MAPK (mitogen-activated protein kinase). There is now sufficient evidence that excess salt intake may be related to the development and progression of SLE and RA, although there are still knowledge gaps. More studies are warranted to further highlight the relationship between excess salt intake, SLE, and RA. Salt intake may affect cell types and pro-inflammatory cytokines and signaling pathways associated with the development and progression of systemic lupus erythematosus and rheumatoid arthritis. Bcl-6 B-cell lymphoma, 6 Erk extracellular signal-regulated kinases, IFN-γ interferon-gamma, JNK c-Jun N-terminal kinase, IL-4 interleukin 4, IL-6 interleukin 6, MAPK mitogen-activated protein kinase, STAT signal transducer and activator of transcription, Tnf-α tumor necrosis factor, Treg T regulatory cell.
Collapse
Affiliation(s)
- Baris Afsar
- Department of Nephrology, School of Medicine, Suleyman Demirel University, Isparta, 32260, Turkey.
| | - Rengin Elsurer Afsar
- Department of Nephrology, School of Medicine, Suleyman Demirel University, Isparta, 32260, Turkey
| |
Collapse
|
16
|
Sposito F, Northey S, Charras A, McNamara PS, Hedrich CM. Hypertonic saline induces inflammation in human macrophages through the NLRP1 inflammasome. Genes Immun 2023; 24:263-269. [PMID: 37573430 PMCID: PMC10575766 DOI: 10.1038/s41435-023-00218-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Revised: 07/27/2023] [Accepted: 08/02/2023] [Indexed: 08/14/2023]
Abstract
Nebulized hypertonic saline (3-7%) is commonly used to increase mucociliary clearance in patients with chronic airway disease and/or virus infections. However, altered salt concentrations may contribute to inflammatory responses. The aim of this study was to investigate whether 500 mM NaCl (3%) triggers inflammation in human macrophages and identify the molecular mechanisms involved. NaCl-induced pyroptosis, IL-1β, IL-18 and ASC speck release were measured in primary human monocyte-derived macrophages. Treatment with the recombinant IL-1 receptor antagonist anakinra or the NLRP3 inhibitor MCC950 did not affect NaCl-mediated inflammasome assembly. Knock-down of NLRP1 expression, but not of NLRP3 and NLRC4, reduced NaCl-induced pyroptosis, pro-inflammatory cytokine and ASC speck release from human THP-1-derived macrophages. Data from this study suggest that 3% NaCl-induced inflammatory responses in human macrophages depend on NLRP1 and inflammasome assembly. Targeting inflammation in addition to inhalation with hypertonic saline may benefit patients with inflammatory airway disease.
Collapse
Affiliation(s)
- Francesca Sposito
- Department of Women's & Children's Health, Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool, UK
| | - Sarah Northey
- Department of Women's & Children's Health, Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool, UK
| | - Amandine Charras
- Department of Women's & Children's Health, Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool, UK
| | - Paul S McNamara
- Department of Women's & Children's Health, Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool, UK
- Department of Respiratory Medicine, Alder Hey Children's NHS Foundation Trust Hospital, Liverpool, UK
| | - Christian M Hedrich
- Department of Women's & Children's Health, Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool, UK.
- Department of Paediatric Rheumatology, Alder Hey Children's NHS Foundation Trust Hospital, Liverpool, UK.
| |
Collapse
|
17
|
Padilla N, Fabbri A, Della-Morte D, Ricordi C, Infante M. Papel inmunomodulador de la vitamina D y los ácidos grasos poliinsaturados omega-3 en trastornos autoinmunes: Revisión de la Literatura. ARCHIVOS LATINOAMERICANOS DE NUTRICIÓN 2023; 73:223-232. [DOI: 10.37527/2023.73.3.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/12/2025]
Abstract
Los trastornos autoinmunes representan una familia de al menos 80 condiciones diferentes que surgen de una respuesta aberrante del sistema inmunológico resultando finalmente en la destrucción de tejidos y órganos específicos del cuerpo. Es importante destacar que durante las últimas tres décadas los estudios epidemiológicos han proporcionado evidencia de un aumento constante en la incidencia y prevalencia de trastornos autoinmunes. En los últimos años, varios estudios han demostrado que la vitamina D y los ácidos grasos poliinsaturados (AGPs) omega-3 ejercen propiedades inmunomoduladoras y antiinflamatorias sinérgicas que pueden aprovecharse positivamente para la prevención y el tratamiento de trastornos autoinmunes. En este sentido, el reciente ensayo clínico denominado VITAL (ensayo de vitamina D y omega 3); un estudio a gran escala, aleatorizado, doble ciego, controlado con placebo encontró que la suplementación conjunta de vitamina D y AGPs omega-3 (VIDOM) puede reducir la incidencia de enfermedades autoinmunes. En esta revisión de la literatura, resumimos los mecanismos moleculares detrás de las propiedades inmunomoduladoras y antiinflamatorias de la vitamina D y los AGPs omega-3, así como la posible interacción bidireccional entre el metabolismo de la vitamina D y el metabolismo de los AGPs omega-3 que justifica la co- suplementación VIDOM en trastornos autoinmunes.
Collapse
|
18
|
Guo H, Li L, Liu B, Lu P, Cao Z, Ji X, Li L, Ouyang G, Nie Z, Lyu A, Lu C. Inappropriate treatment response to DMARDs: A pathway to difficult-to-treat rheumatoid arthritis. Int Immunopharmacol 2023; 122:110655. [PMID: 37481847 DOI: 10.1016/j.intimp.2023.110655] [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: 05/14/2023] [Revised: 07/03/2023] [Accepted: 07/12/2023] [Indexed: 07/25/2023]
Abstract
In recent years, difficult-to-treat rheumatoid arthritis (D2T RA) has attracted significant attention from rheumatologists due to its poor treatment response and the persistent symptoms or signs experienced by patients. The therapeutic demands of patients with D2T RA are not properly met due to unclear pathogenic causes and a lack of high-quality data for current treatment options, creating considerable management difficulties with this patient population. This review describes the clinical challenges associated with disease-modifying antirheumatic drugs (DMARDs) and explores contributing factors associated with inappropriate response to DMARDs that may lead to D2T RA and related immunological dysregulation. It is now understood that D2T RA is a highly heterogeneous pathological status that involves multiple factors. These factors include but are not limited to genetics, environment, immunogenicity, comorbidities, adverse drug reactions, inappropriate drug application, poor adherence, and socioeconomic status. Besides, these factors may manifest in the selection and utilization of specific DMARDs, either individually or in combination, thereby contributing to inadequate treatment response. Finding these variables may offer hints for enhancing DMARD therapy plans and bettering the condition of D2T RA patients.
Collapse
Affiliation(s)
- Hongtao Guo
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, 16# Dongzhimen Nei Nan Xiao Jie, Dongcheng District, Beijing 100700, China; Department of Rheumatology, the First Affiliated Hospital of Henan University of TCM, Zhengzhou 450000, China
| | - Li Li
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, 16# Dongzhimen Nei Nan Xiao Jie, Dongcheng District, Beijing 100700, China
| | - Bin Liu
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, 16# Dongzhimen Nei Nan Xiao Jie, Dongcheng District, Beijing 100700, China
| | - Peipei Lu
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, 16# Dongzhimen Nei Nan Xiao Jie, Dongcheng District, Beijing 100700, China
| | - Zhiwen Cao
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, 16# Dongzhimen Nei Nan Xiao Jie, Dongcheng District, Beijing 100700, China
| | - Xinyu Ji
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, 16# Dongzhimen Nei Nan Xiao Jie, Dongcheng District, Beijing 100700, China
| | - Li Li
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, 16# Dongzhimen Nei Nan Xiao Jie, Dongcheng District, Beijing 100700, China
| | - Guilin Ouyang
- Shanghai Guanghua Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai, China
| | - Zhixin Nie
- Shanghai Guanghua Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai, China
| | - Aiping Lyu
- Shanghai Guanghua Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai, China; School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China
| | - Cheng Lu
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, 16# Dongzhimen Nei Nan Xiao Jie, Dongcheng District, Beijing 100700, China.
| |
Collapse
|
19
|
Kim JY, Lee S, Jang S, Kim CW, Gu BH, Kim M, Kim I. T helper cell polarity determines salt sensitivity and hypertension development. Hypertens Res 2023; 46:2168-2178. [PMID: 37463980 DOI: 10.1038/s41440-023-01365-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Revised: 06/11/2023] [Accepted: 06/15/2023] [Indexed: 07/20/2023]
Abstract
High-salt intake is known to induce pathogenic T helper (Th) 17 cells and hypertension, but contrary to what is known, causes hypertension only in salt-sensitive (SS) individuals. Thus, we hypothesized that Th cell polarity determines salt sensitivity and hypertension development. Cultured splenic T cells from Dahl SS and salt-resistant (SR) rats subjected to hypertonic salt solutions were evaluated via ELISA, flow cytometry, immunocytochemistry and RT-qPCR. Seven-week-old SS and SR rats were fed a chow (CD) or high-salt diet (HSD) for 4 weeks, with weekly measurements of systolic blood pressure. The relaxation response of the aorta rings to the cumulative addition of acetylcholine was measured ex vivo. In these experimental animals, the Th cell polarity (Th17 and T regulatory [Treg]), the expression of Th17- or Treg-related genes, and the enrichment of the transcription factors RORγt and FOXP3 on the target gene promoter regions were determined via flow cytometry, RT-qPCR, and chromatin immunoprecipitation. Hypertonic salt solution induced Th17 and Treg cell differentiation in cultured splenic T cells isolated from SS and SR rats, respectively. HSD induced hypertension, endothelial dysfunction and proinflammatory Th17 cell differentiation only in SS rats. The enrichment of RORγt on the promoter regions of Il17a and Il23r increased their expression only in SS rats. Regardless of HSD, SR rats remained normotensive with Treg polarity, causing high Treg-related gene expressions (Il10, Cd25 and Foxp3). This study demonstrated that Th cell polarity determines salt sensitivity and drives hypertension development. SR rats were protected from HSD-associated hypertension via anti-inflammatory Treg polarity.
Collapse
Affiliation(s)
- Jee Young Kim
- Department of Pharmacology, Kyungpook National University, Daegu, 41944, Republic of Korea
- Cardiovascular Research Institute, Kyungpook National University, Daegu, 41944, Republic of Korea
- BK21 Plus KNU Biomedical Convergence Program, Kyungpook National University, Daegu, 41944, Republic of Korea
- Department of Biomedical Science, School of Medicine, Kyungpook National University, Daegu, 41944, Republic of Korea
| | - Soyung Lee
- Department of Pharmacology, Kyungpook National University, Daegu, 41944, Republic of Korea
- Cardiovascular Research Institute, Kyungpook National University, Daegu, 41944, Republic of Korea
- BK21 Plus KNU Biomedical Convergence Program, Kyungpook National University, Daegu, 41944, Republic of Korea
- Department of Biomedical Science, School of Medicine, Kyungpook National University, Daegu, 41944, Republic of Korea
| | - Sungmin Jang
- Department of Pharmacology, Kyungpook National University, Daegu, 41944, Republic of Korea
- Cardiovascular Research Institute, Kyungpook National University, Daegu, 41944, Republic of Korea
- BK21 Plus KNU Biomedical Convergence Program, Kyungpook National University, Daegu, 41944, Republic of Korea
- Department of Biomedical Science, School of Medicine, Kyungpook National University, Daegu, 41944, Republic of Korea
| | - Cheong-Wun Kim
- Department of Pharmacology, Kyungpook National University, Daegu, 41944, Republic of Korea
- Cardiovascular Research Institute, Kyungpook National University, Daegu, 41944, Republic of Korea
- BK21 Plus KNU Biomedical Convergence Program, Kyungpook National University, Daegu, 41944, Republic of Korea
- Department of Biomedical Science, School of Medicine, Kyungpook National University, Daegu, 41944, Republic of Korea
| | - Bon-Hee Gu
- Department of Animal Science, College of Natural Resources & Live Science, Pusan National University, Miryang, 50463, Republic of Korea
- Life and Industry Convergence Research Institute, Pusan National University, Miryang, 50463, Republic of Korea
| | - Myunghoo Kim
- Department of Animal Science, College of Natural Resources & Live Science, Pusan National University, Miryang, 50463, Republic of Korea
- Life and Industry Convergence Research Institute, Pusan National University, Miryang, 50463, Republic of Korea
| | - Inkyeom Kim
- Department of Pharmacology, Kyungpook National University, Daegu, 41944, Republic of Korea.
- Cardiovascular Research Institute, Kyungpook National University, Daegu, 41944, Republic of Korea.
- BK21 Plus KNU Biomedical Convergence Program, Kyungpook National University, Daegu, 41944, Republic of Korea.
- Department of Biomedical Science, School of Medicine, Kyungpook National University, Daegu, 41944, Republic of Korea.
| |
Collapse
|
20
|
Zlatar L, Mahajan A, Muñoz-Becerra M, Weidner D, Bila G, Bilyy R, Titze J, Hoffmann MH, Schett G, Herrmann M, Steffen U, Muñoz LE, Knopf J. Suppression of neutrophils by sodium exacerbates oxidative stress and arthritis. Front Immunol 2023; 14:1174537. [PMID: 37600805 PMCID: PMC10433750 DOI: 10.3389/fimmu.2023.1174537] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Accepted: 07/14/2023] [Indexed: 08/22/2023] Open
Abstract
Introduction Typical Western diet, rich in salt, contributes to autoimmune disease development. However, conflicting reports exist about the effect of salt on neutrophil effector functions, also in the context of arthritis. Methods We investigated the effect of sodium chloride (NaCl) on neutrophil viability and functions in vitro, and in vivo employing the murine K/BxN-serum transfer arthritis (STA) model. Results and discussion The effects of NaCl and external reactive oxygen species (H2O2) were further examined on osteoclasts in vitro. Hypertonic sodium-rich media caused primary/secondary cell necrosis, altered the nuclear morphology, inhibited phagocytosis, degranulation, myeloperoxidase (MPO) peroxidation activity and neutrophil extracellular trap (NET) formation, while increasing total ROS production, mitochondrial ROS production, and neutrophil elastase (NE) activity. High salt diet (HSD) aggravated arthritis by increasing inflammation, bone erosion, and osteoclast differentiation, accompanied by increased NE expression and activity. Osteoclast differentiation was decreased with 25 mM NaCl or 100 nM H2O2 addition to isotonic media. In contrast to NaCl, external H2O2 had pro-resorptive effects in vitro. We postulate that in arthritis under HSD, increased bone erosion can be attributed to an enhanced oxidative milieu maintained by infiltrating neutrophils, rather than a direct effect of NaCl.
Collapse
Affiliation(s)
- Leticija Zlatar
- Department of Internal Medicine 3 – Rheumatology and Immunology, Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany
- Deutsches Zentrum für Immuntherapie (DZI), Friedrich-Alexander-University Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Aparna Mahajan
- Department of Internal Medicine 3 – Rheumatology and Immunology, Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany
- Deutsches Zentrum für Immuntherapie (DZI), Friedrich-Alexander-University Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Marco Muñoz-Becerra
- Department of Internal Medicine 3 – Rheumatology and Immunology, Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany
- Deutsches Zentrum für Immuntherapie (DZI), Friedrich-Alexander-University Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Daniela Weidner
- Department of Internal Medicine 3 – Rheumatology and Immunology, Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Galyna Bila
- Department of Histology, Cytology, Embryology, Danylo Halytsky Lviv National Medical University, Lviv, Ukraine
| | - Rostyslav Bilyy
- Department of Histology, Cytology, Embryology, Danylo Halytsky Lviv National Medical University, Lviv, Ukraine
| | - Jens Titze
- Division of Nephrology and Hypertension, Universitätsklinikum Erlangen, Erlangen, Germany
- Programme in Cardiovascular and Metabolic Disorders, Duke-NUS Medical School, Singapore, Singapore
| | - Markus H. Hoffmann
- Department of Dermatology, Allergology, and Venereology, University of Lübeck, Lübeck, Germany
| | - Georg Schett
- Department of Internal Medicine 3 – Rheumatology and Immunology, Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany
- Deutsches Zentrum für Immuntherapie (DZI), Friedrich-Alexander-University Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Martin Herrmann
- Department of Internal Medicine 3 – Rheumatology and Immunology, Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany
- Deutsches Zentrum für Immuntherapie (DZI), Friedrich-Alexander-University Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Ulrike Steffen
- Department of Internal Medicine 3 – Rheumatology and Immunology, Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany
- Deutsches Zentrum für Immuntherapie (DZI), Friedrich-Alexander-University Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Luis E. Muñoz
- Department of Internal Medicine 3 – Rheumatology and Immunology, Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany
- Deutsches Zentrum für Immuntherapie (DZI), Friedrich-Alexander-University Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Jasmin Knopf
- Department of Internal Medicine 3 – Rheumatology and Immunology, Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany
- Deutsches Zentrum für Immuntherapie (DZI), Friedrich-Alexander-University Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany
- Department of Pediatric Surgery, University Medical Center Mannheim, University of Heidelberg, Mannheim, Germany
| |
Collapse
|
21
|
Jeong SJ, Yang HJ, Yang HG, Ryu MS, Ha G, Jeong DY, Park S. Inverse association of daily fermented soybean paste (“Jang”) intake with metabolic syndrome risk, especially body fat and hypertension, in men of a large hospital-based cohort. Front Nutr 2023; 10:1122945. [PMID: 36992908 PMCID: PMC10040601 DOI: 10.3389/fnut.2023.1122945] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Accepted: 02/24/2023] [Indexed: 03/16/2023] Open
Abstract
IntroductionJang is a fermented soybean paste containing salt and is traditionally used as a substitute for salt to enhance the flavor of foods in Korea. It has been speculated that regular consumption of Jang may lower the risk of metabolic syndrome (MetS). We hypothesized that Jang intake was associated with the risk of MetS and its components after adjusting for potential confounders, including sodium intake. The hypothesis was investigated according to gender in a large city hospital-based cohort (n = 58,701) in Korea.MethodsJang intake, calculated as the sum of the intakes of Chungkookjang, Doenjang, Doenjang soup, and Ssamjang (a mixture of Doenjang and Kochujang), was included in the semi-quantitative food frequency questionnaire (SQFFQ) administered to the cohort, and the daily Jang intake was estimated. The participants were categorized into low-Jang and high-Jang groups by 1.9 g daily Jang intake. MetS was defined according to 2005 revised United States National Cholesterol Education Program-Adult Treatment Panel III (NCEP-ATP III) criteria modified for Asians.ResultsThe participants in the low-Jang and high-Jang groups consumed an average of 0.63 g and 4.63 g Jang daily; their total sodium intake was about 1.91 and 2.58 g/day, respectively. The participants in the high-Jang group had higher energy, fiber, calcium, vitamin C, vitamin D, and potassium intake than those in the low-Jang group. After adjusting for covariates, the participants with the highest sodium intake (≥3.31 g/day) were positively associated with MetS risk in the quintiles of men and women. Among the MetS components, waist circumference, fat mass, and hypo-high-density lipoprotein (HDL)-cholesterolemia were positively associated with sodium intake in all participants and women. Unlike the association seen with sodium intake, Jang intake (≥1.9 g/day) was inversely associated with MetS components, including waist circumference, fat mass, serum glucose concentrations, and hypo-HDL-cholesterolemia in all participants and men, after adjusting for covariates including sodium intake.DiscussionSubstituting salt for Jang in cooking may be recommended to prevent and alleviate MetS incidence, and its efficacy for MetS risk was better in men than women. The results can be applied to sodium intake in Asian countries where salt is used to promote flavor.
Collapse
Affiliation(s)
- Su-Ji Jeong
- Department of R&D, Sunchang Research Center for Fermentation Microbes, Sunchang-Gun, Republic of Korea
| | - Hee-Jong Yang
- Department of R&D, Sunchang Research Center for Fermentation Microbes, Sunchang-Gun, Republic of Korea
| | - Hee Gun Yang
- Department of R&D, Sunchang Research Center for Fermentation Microbes, Sunchang-Gun, Republic of Korea
| | - Myeong Seon Ryu
- Department of R&D, Sunchang Research Center for Fermentation Microbes, Sunchang-Gun, Republic of Korea
| | - Gwangsu Ha
- Department of R&D, Sunchang Research Center for Fermentation Microbes, Sunchang-Gun, Republic of Korea
| | - Do Yeon Jeong
- Department of R&D, Sunchang Research Center for Fermentation Microbes, Sunchang-Gun, Republic of Korea
- *Correspondence: Do Yeon Jeong,
| | - Sunmin Park
- Department of Food and Nutrition, Obesity/Diabetes Research Center, Hoseo University, Asan, Republic of Korea
- Department of Bioconvergence, Hoseo University, Asan, Republic of Korea
- Sunmin Park,
| |
Collapse
|
22
|
Dey M, Ganguly A, Dahanukar A. An inhibitory mechanism for suppressing high salt intake in Drosophila. Chem Senses 2023; 48:bjad014. [PMID: 37201555 PMCID: PMC10413321 DOI: 10.1093/chemse/bjad014] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Indexed: 05/20/2023] Open
Abstract
High concentrations of dietary salt are harmful to health. Like most animals, Drosophila melanogaster are attracted to foods that have low concentrations of salt, but show strong taste avoidance of high salt foods. Salt in known on multiple classes of taste neurons, activating Gr64f sweet-sensing neurons that drive food acceptance and 2 others (Gr66a bitter and Ppk23 high salt) that drive food rejection. Here we find that NaCl elicits a bimodal dose-dependent response in Gr64f taste neurons, which show high activity with low salt and depressed activity with high salt. High salt also inhibits the sugar response of Gr64f neurons, and this action is independent of the neuron's taste response to salt. Consistent with the electrophysiological analysis, feeding suppression in the presence of salt correlates with inhibition of Gr64f neuron activity, and remains if high salt taste neurons are genetically silenced. Other salts such as Na2SO4, KCl, MgSO4, CaCl2, and FeCl3 act on sugar response and feeding behavior in the same way. A comparison of the effects of various salts suggests that inhibition is dictated by the cationic moiety rather than the anionic component of the salt. Notably, high salt-dependent inhibition is not observed in Gr66a neurons-response to a canonical bitter tastant, denatonium, is not altered by high salt. Overall, this study characterizes a mechanism in appetitive Gr64f neurons that can deter ingestion of potentially harmful salts.
Collapse
Affiliation(s)
- Manali Dey
- Interdepartmental Neuroscience Program, University of California, Riverside, CA 92521, United States
| | - Anindya Ganguly
- Interdepartmental Neuroscience Program, University of California, Riverside, CA 92521, United States
| | - Anupama Dahanukar
- Interdepartmental Neuroscience Program, University of California, Riverside, CA 92521, United States
- Department of Molecular, Cell & Systems Biology, University of California, Riverside, CA 92521, United States
| |
Collapse
|
23
|
High K + intake alleviates experimental autoimmune encephalomyelitis (EAE) and increases T regulatory cells. Cell Immunol 2022; 382:104637. [PMID: 36343517 DOI: 10.1016/j.cellimm.2022.104637] [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: 06/07/2022] [Revised: 10/25/2022] [Accepted: 10/29/2022] [Indexed: 11/06/2022]
Abstract
Multiple sclerosis is believed to be triggered by the interplay between the environmental and genetic factors. In contrast to the Paleolithic diet, the modern Western diet is high in Na+ and low in K+. The present study was undertaken to determine whether high K+ intake alleviated experimental autoimmune encephalomyelitis (EAE), a model of multiple sclerosis. Treatment of C57BL/6 or SJL mice for 7 days with a 5 % K+ diet prior to induction of EAE and maintaining mice on the diet until the end of experiments delayed the onset, reduced the peak, and accelerated the recovery of EAE in both strains compared with mice on a control diet (0.7 % K+), whereas feeding C57BL/6 mice with a 0.1 % K+ diet did the opposite. High K+ intake increased the splenic Treg cell frequency in the pretreatment and peak EAE. Thus, high K+ intake attenuates EAE, possibly by increasing the Treg cells.
Collapse
|
24
|
Chen C, Wang P, Zhang RD, Fang Y, Jiang LQ, Fang X, Zhao Y, Wang DG, Ni J, Pan HF. Mendelian randomization as a tool to gain insights into the mosaic causes of autoimmune diseases. Clin Exp Rheumatol 2022; 21:103210. [PMID: 36273526 DOI: 10.1016/j.autrev.2022.103210] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2022] [Revised: 10/06/2022] [Accepted: 10/11/2022] [Indexed: 12/14/2022]
Abstract
Autoimmune diseases (ADs) are a broad range of disorders which are characterized by long-term inflammation and tissue damage arising from an immune response against one's own tissues. It is now widely accepted that the causes of ADs include environmental factors, genetic susceptibility and immune dysregulation. However, the exact etiology of ADs has not been fully elucidated to date. Because observational studies are plagued by confounding factors and reverse causality, no firm conclusions can be drawn about the etiology of ADs. Over the years, Mendelian randomization (MR) analysis has come into focus, offering unique perspectives and insights into the etiology of ADs and promising the discovery of potential therapeutic interventions. In MR analysis, genetic variation (alleles are randomly dispensed during meiosis, usually irrespective of environmental or lifestyle factors) is used instead of modifiable exposure to explore the link between exposure factors and disease or other outcomes. Therefore, MR analysis can provide a valuable method for exploring the causal relationship between different risk factors and ADs when its inherent assumptions and limitations are fully considered. This review summarized the recent findings of MR in major ADs, including systemic lupus erythematosus (SLE), rheumatoid arthritis (RA), multiple sclerosis (MS), and type 1 diabetes mellitus (T1DM), focused on the effects of different risk factors on ADs risks. In addition, we also discussed the opportunities and challenges of MR methods in ADs research.
Collapse
Affiliation(s)
- Cong Chen
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, 81 Meishan Road, Hefei 230032, Anhui, China; Institute of Kidney Disease, Inflammation & Immunity Mediated Diseases, The Second Hospital of Anhui Medical University, China
| | - Peng Wang
- Institute of Kidney Disease, Inflammation & Immunity Mediated Diseases, The Second Hospital of Anhui Medical University, China; Teaching Center for Preventive Medicine, School of Public Health, Anhui Medical University, 81 Meishan Road, Hefei 230032, Anhui, China
| | - Ruo-Di Zhang
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, 81 Meishan Road, Hefei 230032, Anhui, China; Institute of Kidney Disease, Inflammation & Immunity Mediated Diseases, The Second Hospital of Anhui Medical University, China
| | - Yang Fang
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, 81 Meishan Road, Hefei 230032, Anhui, China; Institute of Kidney Disease, Inflammation & Immunity Mediated Diseases, The Second Hospital of Anhui Medical University, China
| | - Ling-Qiong Jiang
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, 81 Meishan Road, Hefei 230032, Anhui, China; Institute of Kidney Disease, Inflammation & Immunity Mediated Diseases, The Second Hospital of Anhui Medical University, China
| | - Xi Fang
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, 81 Meishan Road, Hefei 230032, Anhui, China; Institute of Kidney Disease, Inflammation & Immunity Mediated Diseases, The Second Hospital of Anhui Medical University, China
| | - Yan Zhao
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, 81 Meishan Road, Hefei 230032, Anhui, China; Institute of Kidney Disease, Inflammation & Immunity Mediated Diseases, The Second Hospital of Anhui Medical University, China
| | - De-Guang Wang
- Institute of Kidney Disease, Inflammation & Immunity Mediated Diseases, The Second Hospital of Anhui Medical University, China; Department of Nephrology, The Second Hospital of Anhui Medical University, Hefei, China.
| | - Jing Ni
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, 81 Meishan Road, Hefei 230032, Anhui, China; Institute of Kidney Disease, Inflammation & Immunity Mediated Diseases, The Second Hospital of Anhui Medical University, China.
| | - Hai-Feng Pan
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, 81 Meishan Road, Hefei 230032, Anhui, China; Institute of Kidney Disease, Inflammation & Immunity Mediated Diseases, The Second Hospital of Anhui Medical University, China.
| |
Collapse
|
25
|
Krajina I, Stupin A, Šola M, Mihalj M. Oxidative Stress Induced by High Salt Diet—Possible Implications for Development and Clinical Manifestation of Cutaneous Inflammation and Endothelial Dysfunction in Psoriasis vulgaris. Antioxidants (Basel) 2022; 11:antiox11071269. [PMID: 35883760 PMCID: PMC9311978 DOI: 10.3390/antiox11071269] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Revised: 06/19/2022] [Accepted: 06/23/2022] [Indexed: 02/07/2023] Open
Abstract
Although oxidative stress is recognized as an important effector mechanism of the immune system, uncontrolled formation of reactive oxygen and nitrogen species promotes excessive tissue damage and leads to disease development. In view of this, increased dietary salt intake has been found to damage redox systems in the vessel wall, resulting in endothelial dysfunction associated with NO uncoupling, inflammation, vascular wall remodeling and, eventually, atherosclerosis. Several studies have reported increased systemic oxidative stress accompanied by reduced antioxidant capacity following a high salt diet. In addition, vigorous ionic effects on the immune mechanisms, such as (trans)differentiation of T lymphocytes are emerging, which together with the evidence of NaCl accumulation in certain tissues warrants a re-examination of the data derived from in vitro research, in which the ionic influence was excluded. Psoriasis vulgaris (PV), as a primarily Th17-driven inflammatory skin disease with proven inflammation-induced accumulation of sodium chloride in the skin, merits our interest in the role of oxidative stress in the pathogenesis of PV, as well as in the possible beneficial effects that could be achieved through modulation of dietary salt intake and antioxidant supplementation.
Collapse
Affiliation(s)
- Ivana Krajina
- Department of Dermatology and Venereology, Osijek University Hospital, J. Huttlera 4, HR-31000 Osijek, Croatia;
- Faculty of Medicine, Josip Juraj Strossmayer University of Osijek, J. Huttlera 4, HR-31000 Osijek, Croatia
| | - Ana Stupin
- Scientific Center of Excellence for Personalized Health Care, Josip Juraj Strossmayer University of Osijek, Trg Svetog Trojstva 3, HR-31000 Osijek, Croatia;
- Institute and Department of Physiology and Immunology, Faculty of Medicine, Josip Juraj Strossmayer University of Osijek, J. Huttlera 4, HR-31000 Osijek, Croatia
| | - Marija Šola
- Department of Dermatology and Venereology, Osijek University Hospital, J. Huttlera 4, HR-31000 Osijek, Croatia;
- Faculty of Medicine, Josip Juraj Strossmayer University of Osijek, J. Huttlera 4, HR-31000 Osijek, Croatia
- Correspondence: (M.Š.); (M.M.); Tel.: +385-31-512-800 (M.M.)
| | - Martina Mihalj
- Department of Dermatology and Venereology, Osijek University Hospital, J. Huttlera 4, HR-31000 Osijek, Croatia;
- Scientific Center of Excellence for Personalized Health Care, Josip Juraj Strossmayer University of Osijek, Trg Svetog Trojstva 3, HR-31000 Osijek, Croatia;
- Institute and Department of Physiology and Immunology, Faculty of Medicine, Josip Juraj Strossmayer University of Osijek, J. Huttlera 4, HR-31000 Osijek, Croatia
- Correspondence: (M.Š.); (M.M.); Tel.: +385-31-512-800 (M.M.)
| |
Collapse
|
26
|
NaCl exposure results in increased expression and processing of IL-1β in Meniere's disease patients. Sci Rep 2022; 12:4957. [PMID: 35322136 PMCID: PMC8943007 DOI: 10.1038/s41598-022-08967-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Accepted: 03/03/2022] [Indexed: 11/08/2022] Open
Abstract
Meniere's disease (MD) is a chronic disease that causes episodic vertigo, fluctuating hearing loss, and aural fullness, initially managed by dietary salt reduction, and use of diuretics. Our prior research in autoimmune inner ear disease (AIED) demonstrated that in peripheral blood mononuclear cell (PBMC) from corticosteroid-resistant AIED patients, increased production, processing and release of interleukin-1β (IL-1β) is observed and hearing could be improved with use of anakinra, an interleukin-1 receptor antagonist. We have further identified that in these AIED patients, IL-1β is uniquely processed to a 28 kDa pro-inflammatory product by caspase-7. In the present study, we characterize the production, processing and release of the pro-inflammatory cytokines IL-1β and IL-6 from PBMC of MD (n = 14) patients in response to sodium chloride (NaCl), and determined the effect of the diuretic triamterene-hydrocholothiazide (T-HCTZ), or anakinra in these patients. We observed that PBMC cultured with NaCl from MD patients show processing of IL-1β to the 28 kDa product, and that this product is abrogated with T-HCTZ. Our observations are consistent with other autoimmune diseases where high concentrations of NaCl caused release of pro-inflammatory cytokines and may provide further insight as to the mechanism of disease progression in MD patients.
Collapse
|
27
|
Role of Oxidative Stress in Vascular Low-Grade Inflammation Initiation Due to Acute Salt Loading in Young Healthy Individuals. Antioxidants (Basel) 2022; 11:antiox11030444. [PMID: 35326095 PMCID: PMC8944840 DOI: 10.3390/antiox11030444] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Revised: 02/20/2022] [Accepted: 02/21/2022] [Indexed: 02/01/2023] Open
Abstract
This study aimed to investigate the effect of 7-day high-salt (HS) and the specific role of oxidative stress on vascular low-grade inflammation initiation in young salt-resistant healthy individuals. 30 young healthy individuals adhered to a 7-day low-salt (LS) diet (3.5 g salt/day), followed by a 7-day high-salt (HS) diet (~14.7 g salt/day) protocol. Pro- and anti-inflammatory cytokines, frequencies of peripheral blood Th17 and Treg cells, Th17/Treg ratio, enzymes SGK1, and p38/MAP kinase, as well as biomarkers of endothelial activation and oxidative stress, were measured before and after the 7-day HS diet protocol. Short-term HS diet significantly increased serum level of pro-inflammatory cytokines INF-γ, TNF-α, IL-9, and IL-17A levels, but also of anti-inflammatory cytokines IL-10 and TGF-β1. Relative amount of total SGK1 significantly increased, following the 7-day HS diet. Increased oxidative stress level, following HS diet, was negatively associated with the frequency of Treg cells. The increase in relative amount of total SGK1 in peripheral mononuclear cells following 7-day HS diet suggests lymphocyte (re)activation, in response to HS intake, resulting in enhanced production of pro-inflammatory (IL-17, INF-γ), but also anti-inflammatory cytokines (IL-10 and TGF-β1). Increased oxidative stress, due to HS loading, alters immune regulatory mechanisms, presumably via effects on Treg cells.
Collapse
|
28
|
Zhang D, Pan J, Liu C, Meng F, Zhang Y, Cao J, Cao Y, Zhou H. Identification of sodium homeostasis genes in Camelus bactrianus by whole transcriptome sequencing. FEBS Open Bio 2022; 12:864-876. [PMID: 35147292 PMCID: PMC8972041 DOI: 10.1002/2211-5463.13380] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Revised: 01/25/2022] [Accepted: 02/09/2022] [Indexed: 11/20/2022] Open
Abstract
Salt dietary intake is tightly coupled to human health, and excessive sodium can cause strokes and cardiovascular diseases. Research into the renal medulla of camels exhibiting high salt resistance may aid identification of the mechanisms governing resistance to high salinity. In this study, we used RNA sequencing (RNA‐seq) to show that in the renal medulla of camels under salt stress, 22 mRNAs, 2 long noncoding RNAs (lncRNAs), and 31 microRNAs (miRNAs) exhibited differential expression compared with the free salt‐intake diet group. Using fluorescence in situ hybridization and dual‐luciferase reporter assays, we demonstrated that the lncRNA LNC003834 can bind miRNA‐34a and thereby relieve suppression of the salt‐absorption‐inhibiting SLC14A1 mRNA from miRNA‐34a, suggesting that the above lncRNA‐miRNA‐mRNA act as competing endogenous RNAs (ceRNAs). We subsequently performed short hairpin RNA and small RNA interference and reactive oxygen species (ROS) detection assays to show that SLC6A1, PCBP2, and PEX5L can improve the antioxidation capacity of renal medulla cells of camel by decreasing ROS levels. Our data suggest that camels achieve sodium homeostasis through regulating the expression of salt‐reabsorption‐related genes in the renal medulla, and this involves ceRNAs (SLC14A1 mRNA, LNC003834, and miRNA‐34a) and antioxidant genes (SLC6A1, PCBP2, and PEX5L). These data may assist in the development of treatments for diseases induced by high salt diets.
Collapse
Affiliation(s)
- Dong Zhang
- Inner Mongolia Key Laboratory of Bio-manufacture, Inner Mongolia Agricultural University, Hohhot, China.,College of Life Sciences, Inner Mongolia Agricultural University, Hohhot, China
| | - Jing Pan
- Inner Mongolia Key Laboratory of Bio-manufacture, Inner Mongolia Agricultural University, Hohhot, China.,College of Life Sciences, Inner Mongolia Agricultural University, Hohhot, China.,Department of Reproductive Medicine, Inner Mongolia Maternal and Child Health Care Hospital, Hohhot, China
| | - Chunxia Liu
- Inner Mongolia Key Laboratory of Bio-manufacture, Inner Mongolia Agricultural University, Hohhot, China.,College of Life Sciences, Inner Mongolia Agricultural University, Hohhot, China
| | - Fanhua Meng
- Inner Mongolia Key Laboratory of Bio-manufacture, Inner Mongolia Agricultural University, Hohhot, China.,College of Life Sciences, Inner Mongolia Agricultural University, Hohhot, China
| | - Yanru Zhang
- Inner Mongolia Key Laboratory of Bio-manufacture, Inner Mongolia Agricultural University, Hohhot, China.,College of Life Sciences, Inner Mongolia Agricultural University, Hohhot, China
| | - Junwei Cao
- Inner Mongolia Key Laboratory of Bio-manufacture, Inner Mongolia Agricultural University, Hohhot, China.,College of Life Sciences, Inner Mongolia Agricultural University, Hohhot, China
| | - Yu Cao
- Inner Mongolia Key Laboratory of Bio-manufacture, Inner Mongolia Agricultural University, Hohhot, China.,College of Life Sciences, Inner Mongolia Agricultural University, Hohhot, China.,Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Huanmin Zhou
- Inner Mongolia Key Laboratory of Bio-manufacture, Inner Mongolia Agricultural University, Hohhot, China.,College of Life Sciences, Inner Mongolia Agricultural University, Hohhot, China
| |
Collapse
|
29
|
Correa-Rodríguez M, DelOlmo-Romero S, Pocovi-Gerardino G, Callejas-Rubio JL, Ríos-Fernández R, Ortego-Centeno N, Rueda-Medina B. Dietary Sodium, Potassium, and Sodium to Potassium Ratio in Patients With Systemic Lupus Erythematosus. Biol Res Nurs 2022; 24:235-244. [PMID: 34978207 DOI: 10.1177/10998004211065491] [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] [Indexed: 01/08/2023]
Abstract
Purpose: The aim of this study was to investigate the association between dietary sodium, potassium, and sodium:potassium ratio and clinical disease activity parameters, damage accrual, and cardiovascular disease risk factors in a population of patients with systemic lupus erythematous (SLE). Research design and study sample: A cross-sectional study including a total of 280 patients was conducted (90.4% females; mean age 46.9 ± 12.85 years). Data collection: The SLE Disease Activity Index (SLEDAI-2K) and the SDI Damage Index were used to assess disease activity and disease-related damage, respectively. A 24-hour diet recall was used to estimate dietary intake of sodium and potassium. Results: Dietary sodium intake was significantly associated with anti-dsDNA (β = -.005; 95% CI [.002 .008]; p = .001) and complement C4 level (β = -.002; 95% CI [-.003, .000]; p = .039). Dietary potassium intake was also significantly associated with complement C3 level (β = -.004; 95% CI [-.007, -.001]; p = .021). Multiple logistic regression models revealed a positive association between dietary sodium intake and the risk of having hsCRP > 3 (p = .005) and an inverse association between dietary potassium intake and the risk of having hsCRP > 3 (p = .004). Conclusions: SLE patients with higher dietary sodium and lower dietary potassium intakes had an increased risk of higher hsCRP. Dietary sodium intake was significantly associated with anti-dsDNA and complement C4 level, while dietary potassium intake was associated with complement C3 level, supporting that dietary sodium and potassium intakes might play a key role in markers related to disease activity in SLE patients.
Collapse
Affiliation(s)
- María Correa-Rodríguez
- Institute of Biomedical Research (IBS), Granada, Spain.,Nursing Department, Faculty of Health Sciences, University of Granada, Granada, Spain
| | - Sara DelOlmo-Romero
- Nursing Department, Faculty of Health Sciences, University of Granada, Granada, Spain
| | | | - José-Luis Callejas-Rubio
- Institute of Biomedical Research (IBS), Granada, Spain.,Systemic Autoimmune Diseases Unit, 16581San Cecilio University Hospital, Granada, Spain.,Faculty of Medicine, University of Granada, Granada, Spain
| | - Raquel Ríos-Fernández
- Institute of Biomedical Research (IBS), Granada, Spain.,Systemic Autoimmune Diseases Unit, 16581San Cecilio University Hospital, Granada, Spain.,Faculty of Medicine, University of Granada, Granada, Spain
| | - Norberto Ortego-Centeno
- Institute of Biomedical Research (IBS), Granada, Spain.,Faculty of Medicine, University of Granada, Granada, Spain
| | - Blanca Rueda-Medina
- Institute of Biomedical Research (IBS), Granada, Spain.,Nursing Department, Faculty of Health Sciences, University of Granada, Granada, Spain
| |
Collapse
|
30
|
Balan Y, Packirisamy RM, Mohanraj PS. High dietary salt intake activates inflammatory cascades via Th17 immune cells: impact on health and diseases. Arch Med Sci 2022; 18:459-465. [PMID: 35316907 PMCID: PMC8924833 DOI: 10.5114/aoms.2020.96344] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/06/2019] [Accepted: 01/13/2020] [Indexed: 02/02/2023] Open
Abstract
The incidence of immune-mediated inflammatory diseases (IMIDs) is on the rise. A high salt content in the diet was found to play a crucial role in mediating IMIDs. It was demonstrated that increased salt concentration favors the differentiation of CD4+ cells to pathogenic Th17 cells, which predispose to several inflammatory diseases by modulating the immunological milieu. In auto-immune diseases increased salt concentration causes stable induction of Th17 cells. In cancer, increased salt concentration triggers chronic inflammation and increases vascular endothelial growth factor levels. Salt-mediated proliferation of Th17 cells has been found to reduce nitric oxide production in the endothelial cells, leading to hypertension. Increased salt concentration was found to alter the intestinal flora, which favors local inflammation. This review attempts to explain the role of high salt concentration and its molecular pathways in causing IMIDs.
Collapse
Affiliation(s)
- Yuvaraj Balan
- Pondicherry Institute of Medical Sciences, Kalapet, Puducherry, India
| | | | - P S Mohanraj
- All India Institute of Medical Sciences, Gorakhpur, India
| |
Collapse
|
31
|
Wan C, Chen S, Zhao K, Ren Z, Peng L, Xia H, Wei H, Yu B. Serum Untargeted Metabolism Reveals the Mechanism of L. plantarum ZDY2013 in Alleviating Kidney Injury Induced by High-Salt Diet. Nutrients 2021; 13:nu13113920. [PMID: 34836175 PMCID: PMC8620752 DOI: 10.3390/nu13113920] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2021] [Revised: 10/28/2021] [Accepted: 10/31/2021] [Indexed: 12/24/2022] Open
Abstract
A high-salt diet (HSD) is one of the key risk factors for hypertension and kidney injury. In this study, a HSD C57BL/6J mice model was established with 4% NaCl, and then different concentrations of Lactobacillus plantarum ZDY2013 were intragastrically administered for 2 weeks to alleviate HSD-induced renal injury. For the study, 16S rRNA gene sequencing, non-targeted metabonomics, real-time fluorescent quantitative PCR, and Masson’s staining were used to investigate the mechanism of L. plantarum ZDY2013 in alleviating renal damage. Results showed that HSD caused intestinal inflammation and changed the intestinal permeability of mice, disrupted the balance of intestinal flora, and increased toxic metabolites (tetrahydrocorticosteron (THB), 3-methyhistidine (3-MH), creatinine, urea, and L-kynurenine), resulting in serious kidney damage. Interestingly, L. plantarum ZDY2013 contributed to reconstructing the intestinal flora of mice by increasing the level of Lactobacillus and Bifidobacterium and decreasing that of Prevotella and Bacteroides. Moreover, the reconstructed intestinal microbiota significantly changed the concentration of the metabolites of hosts through metabolic pathways, including TCA cycle, ABC transport, purine metabolism, and histidine metabolism. The content of uremic toxins such as L-kynurenine, creatinine, and urea in the serum of mice was found to be decreased by L. plantarum ZDY2013, which resulted in renal injury alleviation. Our data suggest that L. plantarum ZDY2013 can indeed improve chronic kidney injury by regulating intestinal flora, strengthening the intestinal barrier, limiting inflammatory response, and reducing uremic toxins.
Collapse
Affiliation(s)
- Cuixiang Wan
- State Key Laboratory of Food Science and Technology, Nanchang University, Nanchang 330047, China; (C.W.); (S.C.); (K.Z.); (Z.R.); (L.P.); (H.W.)
- Sino-German Joint Research Institute, Nanchang University, Nanchang 330047, China;
| | - Shufang Chen
- State Key Laboratory of Food Science and Technology, Nanchang University, Nanchang 330047, China; (C.W.); (S.C.); (K.Z.); (Z.R.); (L.P.); (H.W.)
| | - Kui Zhao
- State Key Laboratory of Food Science and Technology, Nanchang University, Nanchang 330047, China; (C.W.); (S.C.); (K.Z.); (Z.R.); (L.P.); (H.W.)
- Sino-German Joint Research Institute, Nanchang University, Nanchang 330047, China;
| | - Zhongyue Ren
- State Key Laboratory of Food Science and Technology, Nanchang University, Nanchang 330047, China; (C.W.); (S.C.); (K.Z.); (Z.R.); (L.P.); (H.W.)
| | - Lingling Peng
- State Key Laboratory of Food Science and Technology, Nanchang University, Nanchang 330047, China; (C.W.); (S.C.); (K.Z.); (Z.R.); (L.P.); (H.W.)
| | - Huiling Xia
- Sino-German Joint Research Institute, Nanchang University, Nanchang 330047, China;
| | - Hua Wei
- State Key Laboratory of Food Science and Technology, Nanchang University, Nanchang 330047, China; (C.W.); (S.C.); (K.Z.); (Z.R.); (L.P.); (H.W.)
- Sino-German Joint Research Institute, Nanchang University, Nanchang 330047, China;
| | - Bo Yu
- Sino-German Joint Research Institute, Nanchang University, Nanchang 330047, China;
- Correspondence: ; Tel.: +86-791-8833-4578
| |
Collapse
|
32
|
Liu Z, Li SK, Huang CK, Huang CF. A High-Sodium Diet Modulates the Immune Response of Food Allergy in a Murine Model. Nutrients 2021; 13:nu13113684. [PMID: 34835940 PMCID: PMC8621805 DOI: 10.3390/nu13113684] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2021] [Revised: 10/13/2021] [Accepted: 10/14/2021] [Indexed: 12/13/2022] Open
Abstract
Mounting evidence demonstrates that a high-salt diet (HSD) not only affects hemodynamic changes but also disrupts immune homeostasis. The T helper 17 (Th17) and regulatory T cells (Tregs) are susceptible to hypersalinity. However, research on the influence of sodium on Th2-mediated food allergies remains scarce. We aimed to investigate the effect of dietary sodium on the immune response to food allergies. Mice maintained on an HSD (4% NaCl), low-salt diet (LSD; 0.4% NaCl), or control diet (CTRL; 1.0% NaCl) were orally sensitized with ovalbumin (OVA) and a cholera toxin (CT) adjuvant, and then subjected to an intragastric OVA challenge. OVA-specific immunoglobulin G (IgG), IgG1, IgG2a, and IgE antibodies were significantly higher in the HSD group than in the CTRL group (p < 0.001, p < 0.05, p < 0.01, and p < 0.05, respectively). Mice on HSD had significantly higher interleukin (IL)-4 levels than the CTRL group (p < 0.01). The IL-10 levels were significantly lower in the HSD group than in the CTRL group (p < 0.05). The serum levels of interferon-γ (IFN-γ), sodium, and chloride did not differ among the three groups. This study indicates that excessive salt intake promotes Th2 responses in a mouse model of food allergy.
Collapse
Affiliation(s)
- Zheying Liu
- Emergency Department, Department of Emergency and Critical Medicine, Wan Fang Hospital, Taipei Medical University, Taipei City 11696, Taiwan;
- Department of Emergency, School of Medicine, College of Medicine, Taipei Medical University, Taipei City 11031, Taiwan
- Department of Pediatrics, Wan Fang Hospital, Taipei Medical University, Taipei City 11696, Taiwan
- Division of Pediatric Gastroenterology, Hepatology and Nutrition, Department of Pediatrics, Taipei Veterans General Hospital, Taipei City 11217, Taiwan; (S.-K.L.); (C.-K.H.)
| | - Shih-Kuan Li
- Division of Pediatric Gastroenterology, Hepatology and Nutrition, Department of Pediatrics, Taipei Veterans General Hospital, Taipei City 11217, Taiwan; (S.-K.L.); (C.-K.H.)
- Department of Pediatrics, Yonghe Cardinal Tien Hospital, New Taipei City 23445, Taiwan
| | - Chih-Kang Huang
- Division of Pediatric Gastroenterology, Hepatology and Nutrition, Department of Pediatrics, Taipei Veterans General Hospital, Taipei City 11217, Taiwan; (S.-K.L.); (C.-K.H.)
- Department of Pediatrics, Taipei Veterans General Hospital, Taoyuan Branch, Taoyuan City 33052, Taiwan
| | - Ching-Feng Huang
- Division of Pediatric Gastroenterology, Hepatology and Nutrition, Department of Pediatrics, Taipei Veterans General Hospital, Taipei City 11217, Taiwan; (S.-K.L.); (C.-K.H.)
- National Defense Medical Center, School of Medicine, Taipei City 11490, Taiwan
- Correspondence: ; Tel.: +886-2-2875-7019; Fax: +886-2-2873-9019
| |
Collapse
|
33
|
Minamino H, Katsushima M, Hashimoto M, Fujita Y, Yoshida T, Ikeda K, Isomura N, Oguri Y, Yamamoto W, Watanabe R, Murakami K, Murata K, Nishitani K, Tanaka M, Ito H, Ohmura K, Matsuda S, Inagaki N, Morinobu A. Urinary sodium-to-potassium ratio associates with hypertension and current disease activity in patients with rheumatoid arthritis: a cross-sectional study. Arthritis Res Ther 2021; 23:96. [PMID: 33773587 PMCID: PMC8004419 DOI: 10.1186/s13075-021-02479-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Accepted: 03/12/2021] [Indexed: 12/21/2022] Open
Abstract
Background Excessive salt intake is thought to exacerbate both development of hypertension and autoimmune diseases in animal models, but the clinical impact of excessive salt in rheumatoid arthritis (RA) patients is still unknown. We performed a cross-sectional study to clarify the associations between salt load index (urinary sodium-to-potassium ratio (Na/K ratio)), current disease activity, and hypertension in an RA population. Methods Three hundred thirty-six participants from our cohort database (KURAMA) were enrolled. We used the spot urine Na/K ratio as a simplified index of salt loading and used the 28-Joint RA Disease Activity Score (DAS28-ESR) as an indicator of current RA disease activity. Using these indicators, we evaluated statistical associations between urinary Na/K ratio, DAS28-ESR, and prevalence of hypertension. Results Urinary Na/K ratio was positively associated with measured systolic and diastolic blood pressure and also with prevalence of hypertension even after covariate adjustment (OR 1.34, p < 0.001). In addition, increased urinary Na/K ratio was significantly and positively correlated with DAS28-ESR in multiple regression analysis (estimate 0.12, p < 0.001), as was also the case in gender-separated and prednisolone-separated sub-analyses. Conclusion Urinary Na/K ratio was independently associated with current disease activity as well as with prevalence of hypertension in RA patients. Thus, dietary modifications such as salt restriction and potassium supplementation should be investigated as a potential candidate for attenuating both disease activity and hypertension in RA patients. Supplementary Information The online version contains supplementary material available at 10.1186/s13075-021-02479-x.
Collapse
Affiliation(s)
- Hiroto Minamino
- Department of Diabetes, Endocrinology and Nutrition, Graduate School of Medicine, Kyoto University, 54 Shogoin, Kawahara-cho, Sakyo-ku, Kyoto-shi, Kyoto, 606-8507, Japan. .,Japan Society for the Promotion of Science, 5-3-1 Kojimachi, Chiyoda-ku, Tokyo, 102-0083, Japan.
| | - Masao Katsushima
- Department of Rheumatology and Clinical Immunology, Graduate School of Medicine, Kyoto University, 54 Shogoin, Kawahara-cho, Sakyo-ku, Kyoto-shi, Kyoto, 606-8507, Japan
| | - Motomu Hashimoto
- Department of Advanced Medicine for Rheumatic Diseases, Graduate School of Medicine, Kyoto University, 54 Shogoin, Kawahara-cho, Sakyo-ku, Kyoto-shi, Kyoto, 606-8507, Japan.
| | - Yoshihito Fujita
- Department of Diabetes, Endocrinology and Nutrition, Graduate School of Medicine, Kyoto University, 54 Shogoin, Kawahara-cho, Sakyo-ku, Kyoto-shi, Kyoto, 606-8507, Japan.
| | - Tamami Yoshida
- Department of Epidemiology for Community Health and Medicine, Kyoto Prefectural University of Medicine, 465 Kajii-cho, Kamigyo-ku, Kyoto, Kyoto-shi, Kyoto, 602-8566, Japan
| | - Kaori Ikeda
- Department of Diabetes, Endocrinology and Nutrition, Graduate School of Medicine, Kyoto University, 54 Shogoin, Kawahara-cho, Sakyo-ku, Kyoto-shi, Kyoto, 606-8507, Japan
| | - Nozomi Isomura
- Department of Diabetes, Endocrinology and Nutrition, Graduate School of Medicine, Kyoto University, 54 Shogoin, Kawahara-cho, Sakyo-ku, Kyoto-shi, Kyoto, 606-8507, Japan
| | - Yasuo Oguri
- Department of Diabetes, Endocrinology and Nutrition, Graduate School of Medicine, Kyoto University, 54 Shogoin, Kawahara-cho, Sakyo-ku, Kyoto-shi, Kyoto, 606-8507, Japan
| | - Wataru Yamamoto
- Department of Health Information Management, Kurashiki Sweet Hospital, 3542-1 Nakasho, Krashiki, Okayama, 710-0016, Japan
| | - Ryu Watanabe
- Department of Advanced Medicine for Rheumatic Diseases, Graduate School of Medicine, Kyoto University, 54 Shogoin, Kawahara-cho, Sakyo-ku, Kyoto-shi, Kyoto, 606-8507, Japan
| | - Kosaku Murakami
- Department of Rheumatology and Clinical Immunology, Graduate School of Medicine, Kyoto University, 54 Shogoin, Kawahara-cho, Sakyo-ku, Kyoto-shi, Kyoto, 606-8507, Japan
| | - Koichi Murata
- Department of Advanced Medicine for Rheumatic Diseases, Graduate School of Medicine, Kyoto University, 54 Shogoin, Kawahara-cho, Sakyo-ku, Kyoto-shi, Kyoto, 606-8507, Japan.,Department of Orthopaedic Surgery, Graduate School of Medicine, Kyoto University, 54 Shogoin, Kawahara-cho, Sakyo-ku, Kyoto-shi, Kyoto, 606-8507, Japan
| | - Kohei Nishitani
- Department of Advanced Medicine for Rheumatic Diseases, Graduate School of Medicine, Kyoto University, 54 Shogoin, Kawahara-cho, Sakyo-ku, Kyoto-shi, Kyoto, 606-8507, Japan
| | - Masao Tanaka
- Department of Advanced Medicine for Rheumatic Diseases, Graduate School of Medicine, Kyoto University, 54 Shogoin, Kawahara-cho, Sakyo-ku, Kyoto-shi, Kyoto, 606-8507, Japan
| | - Hiromu Ito
- Department of Advanced Medicine for Rheumatic Diseases, Graduate School of Medicine, Kyoto University, 54 Shogoin, Kawahara-cho, Sakyo-ku, Kyoto-shi, Kyoto, 606-8507, Japan.,Department of Orthopaedic Surgery, Graduate School of Medicine, Kyoto University, 54 Shogoin, Kawahara-cho, Sakyo-ku, Kyoto-shi, Kyoto, 606-8507, Japan
| | - Koichiro Ohmura
- Department of Rheumatology and Clinical Immunology, Graduate School of Medicine, Kyoto University, 54 Shogoin, Kawahara-cho, Sakyo-ku, Kyoto-shi, Kyoto, 606-8507, Japan
| | - Shuichi Matsuda
- Department of Orthopaedic Surgery, Graduate School of Medicine, Kyoto University, 54 Shogoin, Kawahara-cho, Sakyo-ku, Kyoto-shi, Kyoto, 606-8507, Japan
| | - Nobuya Inagaki
- Department of Diabetes, Endocrinology and Nutrition, Graduate School of Medicine, Kyoto University, 54 Shogoin, Kawahara-cho, Sakyo-ku, Kyoto-shi, Kyoto, 606-8507, Japan
| | - Akio Morinobu
- Department of Rheumatology and Clinical Immunology, Graduate School of Medicine, Kyoto University, 54 Shogoin, Kawahara-cho, Sakyo-ku, Kyoto-shi, Kyoto, 606-8507, Japan
| |
Collapse
|
34
|
Khanna N, Kumar A, Pawar SV. A Review on Rheumatoid Arthritis Interventions and Current Developments. Curr Drug Targets 2021; 22:463-483. [PMID: 33243118 DOI: 10.2174/1389450121999201125200558] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Revised: 09/08/2020] [Accepted: 10/13/2020] [Indexed: 11/22/2022]
Abstract
Rheumatoid arthritis is a chronic autoimmune disorder characterized by inflammation, swelling, and joint destruction primarily affecting the peripheral joints. In recent years, RA has become an alarming concern affecting more than 1.5% of the population worldwide. The majority of the drugs in clinical trials for rheumatoid arthritis are immunomodulatory. The development of novel drugs for RA is impending and scientists are exploring new strategies through various innovative approaches for RA drug development. Treat-to-target and window of opportunity hypothesis are the new approaches that are used to treat, improve outcomes, and prevent long-term use of ineffective therapy, respectively. Novel therapeutic agents (e.g. GM-CSF inhibitors, Matrix metalloproteinase inhibitors) and delivery systems (e.g., Liposomes, Superparamagnetic iron oxide nano particles (SPIONs)) are under investigation for more target based therapy with reduced side effects and toxicity. The new drug discovery and repositioning of previously FDA-approved drugs are also being considered for chronic inflammatory disorder. The review encompasses a vast array of information, including genetics, etiology, clinical symptoms, current treatment, and newer therapeutics approaches, focused on the development of RA interventions. The introduction of the bioinformatics-based approach in RA has also been significantly discussed in the review. This review provides a general understanding of the challenges and uncertainties in the treatment of RA and summarizes the evolving scenario as well as innovative approaches taken into consideration for drug development in rheumatoid arthritis.
Collapse
Affiliation(s)
- Nikita Khanna
- University Institute of Pharmaceutical Sciences (UIPS), Panjab University, Chandigarh, India
| | - Anil Kumar
- University Institute of Pharmaceutical Sciences (UIPS), Panjab University, Chandigarh, India
| | - Sandip V Pawar
- University Institute of Pharmaceutical Sciences (UIPS), Panjab University, Chandigarh, India
| |
Collapse
|
35
|
Sharif K, Kurnick A, Coplan L, Alexander M, Watad A, Amital H, Shoenfeld Y. The Putative Adverse Effects of Bisphenol A on Autoimmune Diseases. Endocr Metab Immune Disord Drug Targets 2021; 22:665-676. [PMID: 33568039 DOI: 10.2174/1871530321666210210154309] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/06/2020] [Revised: 12/11/2020] [Accepted: 12/18/2020] [Indexed: 11/22/2022]
Abstract
Bisphenol A (BPA) is a monomer that is widely used in the manufacture of polycarbonate plastics including storage plastics and baby bottles, and is considered one of the most widely used synthetic compounds in the manufacturing industry. Exposure to BPA mainly occurs after oral ingestion and results from leaks into food and water from plastic containers and according to epidemiological data exposure is widespread and estimated to occur in 90% of individuals. BPA exertspleiotropiceffects and demonstrates estrogen like effects, thus considered an endocrine disrupting chemical. Growing body of evidence highlight the role of BPA in modulating immune responses and signaling pathways resulting in a proinflammatory response by enhancing the differential polarization of immune cells and cytokine production profile to one that is consistent with proinflammation. Indeed, epidemiological studies have uncovered associations between several autoimmune diseases and BPA exposure. Data from animal models provided consistent evidence highlighting the role of BPA in the pathogenesis, exacerbation and perpetuation of various autoimmune phenomena including neuroinflammation in the context of multiple sclerosis, colitis in inflammatory bowel disease, nephritis in systemic lupus erythematosus, and insulitis in type 1 diabetes mellitus. Given the wide spread of BPA use and its effects in immune systemdysregulation, a call for careful assessment of patients' risks and for public health measures are needed to limit exposure and subsequent deleterious effects. The purpose of this paper is to explore the autoimmune triggering mechanisms and present the current literature supporting the role of BPA in the pathogenesis of autoimmune diseases.
Collapse
Affiliation(s)
- Kassem Sharif
- Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv. Israel
| | - Adam Kurnick
- Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv. Israel
| | - Louis Coplan
- Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv. Israel
| | | | - Abdulla Watad
- Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv. Israel
| | - Howard Amital
- Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv. Israel
| | - Yehuda Shoenfeld
- Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv. Israel
| |
Collapse
|
36
|
Matthias J, Heink S, Picard F, Zeiträg J, Kolz A, Chao YY, Soll D, de Almeida GP, Glasmacher E, Jacobsen ID, Riedel T, Peters A, Floess S, Huehn J, Baumjohann D, Huber M, Korn T, Zielinski CE. Salt generates antiinflammatory Th17 cells but amplifies pathogenicity in proinflammatory cytokine microenvironments. J Clin Invest 2021; 130:4587-4600. [PMID: 32484796 DOI: 10.1172/jci137786] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Accepted: 05/14/2020] [Indexed: 12/16/2022] Open
Abstract
Th cells integrate signals from their microenvironment to acquire distinct specialization programs for efficient clearance of diverse pathogens or for immunotolerance. Ionic signals have recently been demonstrated to affect T cell polarization and function. Sodium chloride (NaCl) was proposed to accumulate in peripheral tissues upon dietary intake and to promote autoimmunity via the Th17 cell axis. Here, we demonstrate that high-NaCl conditions induced a stable, pathogen-specific, antiinflammatory Th17 cell fate in human T cells in vitro. The p38/MAPK pathway, involving NFAT5 and SGK1, regulated FoxP3 and IL-17A expression in high-NaCl conditions. The NaCl-induced acquisition of an antiinflammatory Th17 cell fate was confirmed in vivo in an experimental autoimmune encephalomyelitis (EAE) mouse model, which demonstrated strongly reduced disease symptoms upon transfer of T cells polarized in high-NaCl conditions. However, NaCl was coopted to promote murine and human Th17 cell pathogenicity, if T cell stimulation occurred in a proinflammatory and TGF-β-low cytokine microenvironment. Taken together, our findings reveal a context-dependent, dichotomous role for NaCl in shaping Th17 cell pathogenicity. NaCl might therefore prove beneficial for the treatment of chronic inflammatory diseases in combination with cytokine-blocking drugs.
Collapse
Affiliation(s)
- Julia Matthias
- Institute of Virology, Technical University of Munich, Munich, Germany.,German Center for Infection Research, Partner Site Munich, Munich, Germany.,Department of Cellular Immunoregulation, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Sylvia Heink
- Klinikum rechts der Isar, Department of Experimental Neuroimmunology, Technical University of Munich, Munich, Germany.,Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Felix Picard
- Institute for Medical Microbiology and Hygiene, University of Marburg, Marburg, Germany
| | - Julia Zeiträg
- Institute for Immunology, Biomedical Center, Faculty of Medicine, Ludwig Maximilian University of Munich (LMU Munich), Planegg-Martinsried, Germany
| | - Anna Kolz
- Institute of Clinical Neuroimmunology, Hospital and Biomedical Center of LMU Munich, Planegg-Martinsried, Germany
| | - Ying-Yin Chao
- Institute of Virology, Technical University of Munich, Munich, Germany.,German Center for Infection Research, Partner Site Munich, Munich, Germany.,TranslaTUM, Technical University of Munich, Munich, Germany
| | - Dominik Soll
- Institute of Virology, Technical University of Munich, Munich, Germany.,German Center for Infection Research, Partner Site Munich, Munich, Germany
| | - Gustavo P de Almeida
- Institute of Virology, Technical University of Munich, Munich, Germany.,German Center for Infection Research, Partner Site Munich, Munich, Germany.,TranslaTUM, Technical University of Munich, Munich, Germany
| | - Elke Glasmacher
- Roche Innovation Center Munich, pRED, Large Molecule Research, Penzberg, Germany
| | - Ilse D Jacobsen
- Research Group Microbial Immunology, Leibniz Institute for Natural Product Research and Infection Biology - Hans Knöll Institute, Jena, Germany.,Institute of Microbiology, Friedrich Schiller University, Jena, Germany
| | - Thomas Riedel
- Leibniz Institute DSMZ-German Collection of Microorganisms and Cell Cultures, Braunschweig and German Center for Infection Research, Partner Site Hannover-Braunschweig, Hannover-Braunschweig, Germany
| | - Anneli Peters
- Institute of Clinical Neuroimmunology, Hospital and Biomedical Center of LMU Munich, Planegg-Martinsried, Germany
| | - Stefan Floess
- Department of Experimental Immunology, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Jochen Huehn
- Department of Experimental Immunology, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Dirk Baumjohann
- Institute for Immunology, Biomedical Center, Faculty of Medicine, Ludwig Maximilian University of Munich (LMU Munich), Planegg-Martinsried, Germany
| | - Magdalena Huber
- Institute for Medical Microbiology and Hygiene, University of Marburg, Marburg, Germany
| | - Thomas Korn
- Klinikum rechts der Isar, Department of Experimental Neuroimmunology, Technical University of Munich, Munich, Germany.,Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Christina E Zielinski
- Institute of Virology, Technical University of Munich, Munich, Germany.,German Center for Infection Research, Partner Site Munich, Munich, Germany.,Department of Cellular Immunoregulation, Charité - Universitätsmedizin Berlin, Berlin, Germany.,TranslaTUM, Technical University of Munich, Munich, Germany
| |
Collapse
|
37
|
Sun C, Zhou X, Hu Z, Lu W, Zhao Y, Fang Y. Food and salt structure design for salt reducing. INNOV FOOD SCI EMERG 2021. [DOI: 10.1016/j.ifset.2020.102570] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
38
|
Alpízar-Rodríguez D, Finckh A, Gilbert B. The Role of Nutritional Factors and Intestinal Microbiota in Rheumatoid Arthritis Development. Nutrients 2020; 13:nu13010096. [PMID: 33396685 PMCID: PMC7823566 DOI: 10.3390/nu13010096] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2020] [Revised: 12/10/2020] [Accepted: 12/11/2020] [Indexed: 02/07/2023] Open
Abstract
Evidence about the role of nutritional factors and microbiota in autoimmune diseases, and in rheumatoid arthritis (RA) in particular, has grown in recent years, however many controversies remain. The aim of this review is to summarize the role of nutrition and of the intestinal microbiota in the development of RA. We will focus on selected dietary patterns, individual foods and beverages that have been most consistently associated with RA or with the occurrence of systemic autoimmunity associated with RA. We will also review the evidence for a role of the intestinal microbiota in RA development. We propose that diet and digestive microbiota should be considered together in research, as they interact and may both be the target for future preventive interventions in RA.
Collapse
Affiliation(s)
- Deshiré Alpízar-Rodríguez
- Research Unit, Colegio Mexicano de Reumatología, Mexico City 04318, Mexico
- Correspondence: ; Tel.: +52-55-2525-1853
| | - Axel Finckh
- Department of Rheumatology, Geneva University Hospitals, 1206 Geneva, Switzerland; (A.F.); (B.G.)
| | - Benoît Gilbert
- Department of Rheumatology, Geneva University Hospitals, 1206 Geneva, Switzerland; (A.F.); (B.G.)
| |
Collapse
|
39
|
Dent EL, Broome HJ, Sasser JM, Ryan MJ. Blood pressure and albuminuria in a female mouse model of systemic lupus erythematosus: impact of long-term high salt consumption. Am J Physiol Regul Integr Comp Physiol 2020; 319:R448-R454. [PMID: 32813539 DOI: 10.1152/ajpregu.00070.2020] [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] [Indexed: 11/22/2022]
Abstract
Hypertension and kidney involvement are common in patients with autoimmune disease. Sodium intake is linked to hypertension in both human and animal studies. Evidence suggests that dietary salt may be an important environmental factor that promotes autoimmune activity. Therefore, we hypothesized that a long-term high-salt diet would accelerate the progression of autoimmunity, hypertension, and albuminuria during systemic lupus erythematosus (SLE), an autoimmune disease that predominantly affects young women and has a high prevalence of hypertension and renal disease. To test this hypothesis, an established experimental model of SLE (female NZBWF1 mice) that develops hypertension and renal disease was used. SLE mice were fed a high-salt (4% NaCl) or normal (0.4% NaCl) diet for 24 wk beginning at 10 wk of age and ending at 34 wk of age, a time by which female NZBWF1 mice typically have hypertension and exhibit signs of renal disease. Plasma anti-dsDNA autoantibodies were measured as an indicator of active SLE disease, and urinary albumin was monitored longitudinally as a marker of renal disease. Arterial pressure was measured in conscious, freely moving mice at 34 wk of age. Urinary endothelin-1 (ET-1) excretion, renal endothelin A and B receptor protein expression, and renal mRNA expression of NOS1, NOS2, NOX2, MCP-1, TNF-α, serum- and glucocorticoid-regulated kinase 1, and interleukin-2 (IL-2) were assessed to determine the impact on gene products commonly altered by a high-salt diet. SLE mice fed a high-salt diet had increased circulating autoantibodies, but the high-salt diet did not significantly affect albuminuria or arterial pressure. Urinary ET-1 excretion was increased, whereas renal endothelin A receptor and IL-2 expression were decreased in response to a high-salt diet. These data suggest that a chronic high-salt diet may not accelerate cardiovascular and renal consequences commonly associated with SLE.
Collapse
Affiliation(s)
- Elena L Dent
- Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, Mississippi
| | - Hanna J Broome
- Department of Cell and Molecular Biology, University of Mississippi Medical Center, Jackson, Mississippi
| | - Jennifer M Sasser
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, Mississippi
| | - Michael J Ryan
- Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, Mississippi.,G.V (Sonny) Montgomery Veterans Affairs Medical Center, Jackson, Mississippi
| |
Collapse
|
40
|
Yang C, Li J, Sun F, Zhou H, Yang J, Yang C. The functional duality of SGK1 in the regulation of hyperglycemia. Endocr Connect 2020; 9:R187-R194. [PMID: 32621586 PMCID: PMC7424354 DOI: 10.1530/ec-20-0225] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/23/2020] [Accepted: 07/02/2020] [Indexed: 12/11/2022]
Abstract
Hyperglycemia is the consequence of blood glucose dysregulation and a driving force of diabetic complications including retinopathy, nephropathy and cardiovascular diseases. The serum and glucocorticoid inducible kinase-1 (SGK1) has been suggested in the modulation of various pathophysiological activities. However, the role of SGK1 in blood glucose homeostasis remains less appreciated. In this review, we intend to summarize the function of SGK1 in glucose level regulation and to examine the evidence supporting the therapeutic potential of SGK1 inhibitors in hyperglycemia. Ample evidence points to the controversial roles of SGK1 in pancreatic insulin secretion and peripheral insulin sensitivity, which reflects the complex interplay between SGK1 activation and blood glucose fluctuation. Furthermore, SGK1 is engaged in glucose absorption and excretion in intestine and kidney and participates in the progression of hyperglycemia-induced secondary organ damage. As a net effect, blockage of SGK1 activation via either pharmacological inhibition or genetic manipulation seems to be helpful in glucose control at varying diabetic stages.
Collapse
Affiliation(s)
- Chunliang Yang
- Department of Integrated Traditional Chinese and Western Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- The Center for Biomedical Research, Tongji Hospital Research Building, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Junyi Li
- Department of Integrated Traditional Chinese and Western Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Fei Sun
- The Center for Biomedical Research, Tongji Hospital Research Building, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Haifeng Zhou
- Department of Integrated Traditional Chinese and Western Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jia Yang
- Department of Integrated Traditional Chinese and Western Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Correspondence should be addressed to C Yang or J Yang: or
| | - Chao Yang
- Department of Gerontology, Hubei Provincial Hospital of Integrated Chinese and Western Medicine, Wuhan, China
- Correspondence should be addressed to C Yang or J Yang: or
| |
Collapse
|
41
|
Duan X, Tan X, Gu L, Liu J, Hao X, Tao L, Feng H, Cao Y, Shi Z, Duan Y, Deng M, Chen G, Qi C, Zhang Y. New secondary metabolites with immunosuppressive activity from the phytopathogenic fungus Bipolaris maydis. Bioorg Chem 2020; 99:103816. [DOI: 10.1016/j.bioorg.2020.103816] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2019] [Revised: 02/27/2020] [Accepted: 04/02/2020] [Indexed: 11/28/2022]
|
42
|
Mavropoulos A. On the Role of Salt in Immunoregulation and Autoimmunity. Mediterr J Rheumatol 2020; 32:3-5. [PMID: 34386696 PMCID: PMC8314887 DOI: 10.31138/mjr.32.1.3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Accepted: 01/28/2020] [Indexed: 12/19/2022] Open
Affiliation(s)
- Athanasios Mavropoulos
- Department of Rheumatology and Clinical Immunology, Faculty of Medicine, School of Health Sciences, University of Thessaly, Larissa, Greece
| |
Collapse
|
43
|
Le M, Muntyanu A, Netchiporouk E. IncRNAs and circRNAs provide insight into discoid lupus pathogenesis and progression. ANNALS OF TRANSLATIONAL MEDICINE 2020; 8:260. [PMID: 32355704 PMCID: PMC7186711 DOI: 10.21037/atm.2020.03.56] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Affiliation(s)
- Michelle Le
- Division of Dermatology, Department of Medicine, McGill University Health Centre, Montreal, Quebec, Canada
| | - Anastasiya Muntyanu
- Division of Dermatology, Department of Medicine, McGill University Health Centre, Montreal, Quebec, Canada
| | - Elena Netchiporouk
- Division of Dermatology, Department of Medicine, McGill University Health Centre, Montreal, Quebec, Canada
| |
Collapse
|
44
|
Kwiatkowska B, Maślińska M. The place of omega-3 and omega-6 acids in supplementary treatment of inflammatory joint diseases. Reumatologia 2020; 58:34-41. [PMID: 32322122 PMCID: PMC7174795 DOI: 10.5114/reum.2020.93511] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2020] [Accepted: 02/12/2020] [Indexed: 02/06/2023] Open
Abstract
Eating habits have been analysed for years as a factor influencing the development of autoimmune diseases and susceptibility to infections. On the basis of research, observational studies and meta-analyses, special attention was paid to omega-3 and omega-6 acids. The purpose of the review is to show the importance of omega-3 and omega-6 acids as important ingredients in the healthy diet and as factors protecting against the development of the most common inflammatory rheumatic diseases. The influence of these omega-3 and -6 acids on the course of rheumatic diseases and arguments for their use as complementary therapy are also presented.
Collapse
Affiliation(s)
- Brygida Kwiatkowska
- Early Arthritis Clinic, National Institute of Geriatrics, Rheumatology and Rehabilitation, Warsaw, Poland
| | - Maria Maślińska
- Early Arthritis Clinic, National Institute of Geriatrics, Rheumatology and Rehabilitation, Warsaw, Poland
| |
Collapse
|
45
|
Bustamante MF, Agustín-Perez M, Cedola F, Coras R, Narasimhan R, Golshan S, Guma M. Design of an anti-inflammatory diet (ITIS diet) for patients with rheumatoid arthritis. Contemp Clin Trials Commun 2020; 17:100524. [PMID: 32025586 PMCID: PMC6997513 DOI: 10.1016/j.conctc.2020.100524] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2019] [Revised: 01/01/2020] [Accepted: 01/11/2020] [Indexed: 12/17/2022] Open
Abstract
Background Rheumatoid arthritis (RA) is a chronic systemic autoimmune disease that affects synovial joints, leading to inflammation, joint destruction, loss of function, and disability. Although recent pharmaceutical advances have improved treatment of RA, patients with RA often inquire about dietary interventions to improve RA symptoms, as they perceive rapid changes in their symptoms after consumption of certain foods. There is evidence that some ingredients have pro- or anti-inflammatory effects. In addition, recent literature has shown a link between diet and microbiome changes. Both diet and the gut microbiome are linked to circulating metabolites that may modulate inflammation. However, evidence of the effects of an anti-inflammatory and probiotic-rich diet in patients with RA is scarce. There is also a need for biological data to support its anti-inflammatory effects. Methods The main goal of this study is to delineate the design process for a diet tailored to our RA population. To achieve this goal, we collected information on diet, supplements, cooking methods, and intake of different ingredients for each patient. Different groups were interviewed, and their feedback was assessed to design a diet that incorporates suggested anti-inflammatory ingredients in a manner that was easy for patients to adopt based on their lifestyles and backgrounds. Results We designed a diet that includes a high intake of potential anti-inflammatory ingredients. Feedback from highly motivated patients was critical in constructing an anti-inflammatory diet (ITIS diet) with elevated adherence. Conclusion In order to tailor our diet, we surveyed our patients on several different parameters. We obtained important feedback on how feasible our ITIS diet is for RA patients. Using this feedback, we made minor improvements and finalized the design of the ITIS diet. This diet is being used in an on-going pilot study to determine their anti-inflammatory effect in pain and joint swelling in RA patients. Trial registration Not applicable.
Collapse
Affiliation(s)
- Marta F Bustamante
- Department of Medicine, University of California San Diego, San Diego, CA, USA
| | | | - Francesca Cedola
- Department of Medicine, University of California San Diego, San Diego, CA, USA
| | - Roxana Coras
- Department of Medicine, University of California San Diego, San Diego, CA, USA.,Department of Medicine, Autonomous University of Barcelona, Plaça Cívica, 08193, Bellaterra, Barcelona, Spain
| | - Rekha Narasimhan
- Department of Medicine, University of California San Diego, San Diego, CA, USA
| | - Shahrokh Golshan
- Department of Psychiatry, University of California San Diego, San Diego, CA, USA
| | - Monica Guma
- Department of Medicine, University of California San Diego, San Diego, CA, USA.,Department of Medicine, Autonomous University of Barcelona, Plaça Cívica, 08193, Bellaterra, Barcelona, Spain
| |
Collapse
|
46
|
Vojdani A, Gushgari LR, Vojdani E. Interaction between food antigens and the immune system: Association with autoimmune disorders. Autoimmun Rev 2020; 19:102459. [PMID: 31917265 DOI: 10.1016/j.autrev.2020.102459] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2019] [Accepted: 08/19/2019] [Indexed: 02/08/2023]
Abstract
It has been shown that environmental factors such as infections, chemicals, and diet play a major role in autoimmune diseases; however, relatively little attention has been given to food components as the most prevalent modifiers of these afflictions. This review summarizes the current body of knowledge related to different mechanisms and associations between food proteins/peptides and autoimmune disorders. The primary factor controlling food-related immune reactions is the oral tolerance mechanism. The failure of oral tolerance triggers immune reactivity against dietary antigens, which may initiate or exacerbate autoimmune disease when the food antigen shares homology with human tissue antigens. Because the conformational fit between food antigens and a host's self-determinants has been determined for only a few food proteins, we examined evidence related to the reaction of affinity-purified disease-specific antibody with different food antigens. We also studied the reaction of monoclonal or polyclonal tissue-specific antibodies with various food antigens and the reaction of food-specific antibodies with human tissue antigens. Examining the assembled information, we postulated that chemical modification of food proteins by different toxicants in food may result in immune reaction against modified food proteins that cross-react with tissue antigens, resulting in autoimmune reactivity. Because we are what our microbiome eats, food can change the gut commensals, and toxins can breach the gut barrier, penetrating into different organs where they can initiate autoimmune response. Conversely, there are also foods and supplements that help maintain oral tolerance and microbiome homeostasis. Understanding the potential link between specific food consumption and autoimmunity in humans may lay the foundation for further research about the proper diet in the prevention of autoimmune diseases.
Collapse
Affiliation(s)
- Aristo Vojdani
- Immunosciences Lab, Inc., 822 S. Robertson Blvd, Ste. 312, Los Angeles, CA 90035, USA; Department of Preventive Medicine, Loma Linda University, Loma Linda, CA 92350, USA.
| | - Lydia R Gushgari
- Cyrex Laboratories, LLC. 2602 South 24(th) St., Phoenix, AZ 85034, USA.
| | - Elroy Vojdani
- Regenera Medical, 11860 Wilshire Blvd., Ste. 301, Los Angeles, CA 90025, USA.
| |
Collapse
|
47
|
Brito-Zerón P, Acar-Denizli N, Ng WF, Horváth IF, Rasmussen A, Seror R, Li X, Baldini C, Gottenberg JE, Danda D, Quartuccio L, Priori R, Hernandez-Molina G, Armagan B, Kruize AA, Kwok SK, Kvarnstrom M, Praprotnik S, Sene D, Gerli R, Solans R, Rischmueller M, Mandl T, Suzuki Y, Isenberg D, Valim V, Wiland P, Nordmark G, Fraile G, Bootsma H, Nakamura H, Giacomelli R, Devauchelle-Pensec V, Hofauer B, Bombardieri M, Trevisani VFM, Hammenfors D, Pasoto SG, Retamozo S, Gheita TA, Atzeni F, Morel J, Vollenweider C, Zeher M, Sivils K, Xu B, Bombardieri S, Sandhya P, De Vita S, Minniti A, Sánchez-Guerrero J, Kilic L, van der Heijden E, Park SH, Wahren-Herlenius M, Mariette X, Ramos-Casals M. Epidemiological profile and north–south gradient driving baseline systemic involvement of primary Sjögren’s syndrome. Rheumatology (Oxford) 2019; 59:2350-2359. [DOI: 10.1093/rheumatology/kez578] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2019] [Revised: 10/27/2019] [Indexed: 12/21/2022] Open
Abstract
Abstract
Objective
To characterize the systemic phenotype of primary Sjögren’s syndrome at diagnosis by analysing the EULAR-SS disease activity index (ESSDAI) scores.
Methods
The Sjögren Big Data Consortium is an international, multicentre registry based on worldwide data-sharing cooperative merging of pre-existing databases from leading centres in clinical research in Sjögren’s syndrome from the five continents.
Results
The cohort included 10 007 patients (9352 female, mean 53 years) with recorded ESSDAI scores available. At diagnosis, the mean total ESSDAI score was 6.1; 81.8% of patients had systemic activity (ESSDAI score ≥1). Males had a higher mean ESSDAI (8.1 vs 6.0, P < 0.001) compared with females, as did patients diagnosed at <35 years (6.7 vs 5.6 in patients diagnosed at >65 years, P < 0.001). The highest global ESSDAI score was reported in Black/African Americans, followed by White, Asian and Hispanic patients (6.7, 6.5, 5.4 and 4.8, respectively; P < 0.001). The frequency of involvement of each systemic organ also differed between ethnic groups, with Black/African American patients showing the highest frequencies in the lymphadenopathy, articular, peripheral nervous system, CNS and biological domains, White patients in the glandular, cutaneous and muscular domains, Asian patients in the pulmonary, renal and haematological domains and Hispanic patients in the constitutional domain. Systemic activity measured by the ESSDAI, clinical ESSDAI (clinESSDAI) and disease activity states was higher in patients from southern countries (P < 0.001).
Conclusion
The systemic phenotype of primary Sjögren’s syndrome is strongly influenced by personal determinants such as age, gender, ethnicity and place of residence, which are key geoepidemiological players in driving the expression of systemic disease at diagnosis.
Collapse
Affiliation(s)
- Pilar Brito-Zerón
- Department of Medicine, Autoimmune Diseases Unit, Hospital CIMA – Sanitas, Barcelona, Spain
- Sjögren Syndrome Research Group (AGAUR), Laboratory of Autoimmune Diseases Josep Font, IDIBAPS-CELLEX, Department of Autoimmune Diseases, ICMiD, University of Barcelona, Hospital Clínic, Barcelona, Spain
| | - Nihan Acar-Denizli
- Department of Statistics, Faculty of Science and Letters, Mimar Sinan Fine Arts University, Istanbul, Turkey
| | - Wan-Fai Ng
- Institute of Cellular Medicine, Newcastle University, NIHR Biomedical Research Centre, Newcastle Upon Tyne, UK
| | - Ildiko Fanny Horváth
- Division of Clinical Immunology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Astrid Rasmussen
- Arthritis and Clinical Immunology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
| | - Raphaele Seror
- Center for Immunology of Viral Infections and Autoimmune Diseases, Assistance Publique – Hôpitaux de Paris, Hôpitaux Universitaires Paris-Sud, Le Kremlin-Bicêtre, Université Paris Sud, INSERM, Paris, France
| | - Xiaomei Li
- Department of Rheumatology and Immunology, Anhui Provincial Hospital, Hefei, China
| | | | - Jacques-Eric Gottenberg
- Department of Rheumatology, Strasbourg University Hospital, Université de Strasbourg, CNRS, Strasbourg, France
| | - Debashish Danda
- Department of Clinical Immunology & Rheumatology, Christian Medical College & Hospital, Vellore, India
| | - Luca Quartuccio
- Clinic of Rheumatology, Department of Medical Area, University Hospital ‘Santa Maria della Misericordia’, Udine, Italy
| | - Roberta Priori
- Department of Internal Medicine and Medical Specialties, Rheumatology Clinic, Sapienza University of Rome, Rome, Italy
| | - Gabriela Hernandez-Molina
- Immunology and Rheumatology Department, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, México City, Mexico
| | - Berkan Armagan
- Department of Internal Medicine, Hacettepe University, Faculty of Medicine, Ankara, Turkey
| | - Aike A Kruize
- Department of Rheumatology and Clinical Immunology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Seung-Ki Kwok
- Division of Rheumatology, Department of Internal Medicine, College of Medicine, Seoul St Mary's Hospital, The Catholic University of Korea, Seoul, South Korea
| | - Marika Kvarnstrom
- Division of Rheumatology, Department of Medicine, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Sonja Praprotnik
- Department of Rheumatology, University Medical Centre, Ljubljana, Slovenia
| | - Damien Sene
- Département de Médecine Interne, Hôpital Lariboisière, Université Paris VII, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Roberto Gerli
- Rheumatology Unit, Department of Medicine, University of Perugia, Perugia, Italy
| | - Roser Solans
- Department of Internal Medicine, Hospital Vall d'Hebron, Barcelona, Spain
| | - Maureen Rischmueller
- Department of Rheumatology, The Queen Elizabeth Hospital, Discipline of Medicine, University of Adelaide, South Australia, Australia
| | - Thomas Mandl
- Department of Rheumatology, Skane University Hospital Malmö, Lund University, Malmö, Sweden
| | - Yasunori Suzuki
- Division of Rheumatology, Kanazawa University Hospital, Kanazawa, Ishikawa, Japan
| | - David Isenberg
- Centre for Rheumatology, Division of Medicine, University College London, London, UK
| | - Valeria Valim
- Department of Medicine, Federal University of Espírito Santo, Vitória, Brazil
| | - Piotr Wiland
- Department of Rheumatology and Internal Medicine, Wroclaw Medical Hospital, Wroclaw, Poland
| | - Gunnel Nordmark
- Rheumatology, Department of Medical Sciences, University of Uppsala, Uppsala, Sweden
| | - Guadalupe Fraile
- Department of Internal Medicine, Hospital Ramón y Cajal, Madrid, Spain
| | - Hendrika Bootsma
- Department of Rheumatology & Clinical Immunology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Hideki Nakamura
- Department of Immunology and Rheumatology, Division of Advanced Preventive Medical Sciences, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Roberto Giacomelli
- Clinical Unit of Rheumatology, School of Medicine, University of L'Aquila, L'Aquila, Italy
| | | | - Benedikt Hofauer
- Otorhinolaryngology / Head and Neck Surgery, University Medical Center Freiburg, Freiburg, Germany
| | - Michele Bombardieri
- Centre for Experimental Medicine and Rheumatology, Queen Mary University of London, London, UK
| | | | - Daniel Hammenfors
- Section for Rheumatology, Department of Clinical Science, University of Bergen, Bergen, Norway
- Department of Rheumatology, Haukeland University Hospital, Bergen, Norway
| | - Sandra G Pasoto
- Rheumatology Division, Hospital das Clinicas, Faculdade de Medicina da Universidade de Sao Paulo (HCFMUSP), Sao Paulo, Brazil
| | - Soledad Retamozo
- Instituto De Investigaciones En Ciencias De La Salud (INICSA), Universidad Nacional de Córdoba (UNC), Cordoba, Argentina
- Department of Rheumatology, Instituto Modelo de Cariología Privado S.R.L, Instituto Universitario de Ciencias Biomídicas de Córdoba, Cordoba, Argentina
| | - Tamer A Gheita
- Rheumatology Department, Kasr Al Ainy School of Medicine, Cairo University, Cairo, Egypt
| | - Fabiola Atzeni
- IRCCS Galeazzi Orthopedic Institute, Milan, Italy
- Rheumatology Unit, University of Messina, Messina, Italy
| | - Jacques Morel
- Department of Rheumatology, CHU Montpellier, University of Montpellier, Montpellier, France
| | | | - Margit Zeher
- Division of Clinical Immunology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Kathy Sivils
- Arthritis and Clinical Immunology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
| | - Bei Xu
- Department of Rheumatology and Immunology, Anhui Provincial Hospital, Hefei, China
| | | | - Pulukool Sandhya
- Department of Clinical Immunology & Rheumatology, Christian Medical College & Hospital, Vellore, India
| | - Salvatore De Vita
- Clinic of Rheumatology, Department of Medical Area, University Hospital ‘Santa Maria della Misericordia’, Udine, Italy
| | - Antonina Minniti
- Department of Internal Medicine and Medical Specialties, Rheumatology Clinic, Sapienza University of Rome, Rome, Italy
| | - Jorge Sánchez-Guerrero
- Immunology and Rheumatology Department, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, México City, Mexico
| | - Levent Kilic
- Department of Internal Medicine, Hacettepe University, Faculty of Medicine, Ankara, Turkey
| | - Eefje van der Heijden
- Department of Rheumatology and Clinical Immunology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Sung-Hwan Park
- Division of Rheumatology, Department of Internal Medicine, College of Medicine, Seoul St Mary's Hospital, The Catholic University of Korea, Seoul, South Korea
| | - Marie Wahren-Herlenius
- Division of Rheumatology, Department of Medicine, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Xavier Mariette
- Center for Immunology of Viral Infections and Autoimmune Diseases, Assistance Publique – Hôpitaux de Paris, Hôpitaux Universitaires Paris-Sud, Le Kremlin-Bicêtre, Université Paris Sud, INSERM, Paris, France
| | - Manuel Ramos-Casals
- Sjögren Syndrome Research Group (AGAUR), Laboratory of Autoimmune Diseases Josep Font, IDIBAPS-CELLEX, Department of Autoimmune Diseases, ICMiD, University of Barcelona, Hospital Clínic, Barcelona, Spain
| | | |
Collapse
|
48
|
Afroz KF, Alviña K. Maternal elevated salt consumption and the development of autism spectrum disorder in the offspring. J Neuroinflammation 2019; 16:265. [PMID: 31837704 PMCID: PMC6911292 DOI: 10.1186/s12974-019-1666-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2019] [Accepted: 11/27/2019] [Indexed: 01/15/2023] Open
Abstract
Autism spectrum disorder (ASD) is a prevalent neurodevelopmental condition with no known etiology or cure. Several possible contributing factors, both genetic and environmental, are being actively investigated. Amongst these, maternal immune dysregulation has been identified as potentially involved in promoting ASD in the offspring. Indeed, ASD-like behaviors have been observed in studies using the maternal immune activation mouse model. Furthermore, recent studies have shed light on maternal dietary habits and their impact on the gut microbiome as factors possibly facilitating ASD. However, most of these studies have been limited to the effects of high fat and/or high sugar. More recent data, however, have shown that elevated salt consumption has a significant effect on the immune system and gut microbiome, often resulting in gut dysbiosis and induction of pro-inflammatory pathways. Specifically, high salt alters the gut microbiome and induces the differentiation of T helper-17 cells that produce pro-inflammatory cytokines such as interleukin-17 and interleukin-23. Moreover, elevated salt can also reduce the differentiation of regulatory T cells that help maintaining a balanced immune system. While in the innate immune system, high salt can cause over activation of M1 pro-inflammatory macrophages and downregulation of M2 regulatory macrophages. These changes to the immune system are alarming because excessive consumption of salt is a documented worldwide problem. Thus, in this review, we discuss recent findings on high salt intake, gut microbiome, and immune system dysregulation while proposing a hypothesis to link maternal overconsumption of salt and children’s ASD.
Collapse
Affiliation(s)
- Kazi Farhana Afroz
- Department of Biological Sciences, Texas Tech University, 2901 Main St. Room #05, Biology Building, Lubbock, TX, 79409, USA
| | - Karina Alviña
- Department of Biological Sciences, Texas Tech University, 2901 Main St. Room #05, Biology Building, Lubbock, TX, 79409, USA. .,Department of Neuroscience, University of Florida, 1149 Newell Drive, Room L1-100, Gainesville, FL, 32611, USA.
| |
Collapse
|
49
|
Short-Term High-NaCl Dietary Intake Changes Leukocyte Expression of VLA-4, LFA-1, and Mac-1 Integrins in Both Healthy Humans and Sprague-Dawley Rats: A Comparative Study. Mediators Inflamm 2019; 2019:6715275. [PMID: 31636506 PMCID: PMC6766117 DOI: 10.1155/2019/6715275] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2019] [Revised: 07/26/2019] [Accepted: 08/08/2019] [Indexed: 01/11/2023] Open
Abstract
This study is aimed at assessing the effects of a short-term high-salt (HS) diet on the peripheral blood leukocyte (PBL) activation status in healthy rats and young human individuals. Distribution of PBL subpopulations and surface expression of integrins were determined using flow cytometry in 36 men and women on a 7-day low-salt diet (<3.2 g salt/day) immediately followed by a 7-day HS diet (~14 g salt/day) or in Sprague-Dawley (SD) rats (n = 24) on a 0.4% NaCl diet (aLS group) or a 4% NaCl diet (aHS group) for 7 days. The aHS group presented with an increased frequency of granulocytes, while the frequency of lymphocytes was reduced. Although in humans HS diet reduced the expression of CD11b(act) integrin on lymphocytes, the frequency of CD11b(act)-bearing cells among all PBL subsets was increased. The aHS group of rats exhibited increased expression of total CD11b/c in granulocytes and CD3 lymphocytes. The expression of CD11a was significantly reduced in all PBL subsets from human subjects and increased in the aHS group. CD49d expression on all PBL subsets was significantly decreased in both humans and rats. In human subjects, we found reduced frequencies of intermediate monocytes accompanied by a reciprocal increase in classical monocytes. Present results suggest that a short-term HS diet can alter leukocytes' activation status and promote vascular low-grade inflammation.
Collapse
|
50
|
Lima A, Tavares J, Pestana N, Carvalho MJ, Cabrita A, Rodrigues A. Sodium removal in peritoneal dialysis: is there room for a new parameter in dialysis adequacy? BULLETIN DE LA DIALYSE À DOMICILE 2019. [DOI: 10.25796/bdd.v2i3.21343] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022] Open
Abstract
In peritoneal dialysis (PD) (as well as in hemodialysis) small solute clearance measured as Kt/v urea has long been used as a surrogate of dialysis adequacy. A better urea clearance was initially thought to increase survival in dialysis patients (as shown in the CANUSA trial)(1), but reanalysis of the data showed a superior contribution of residual renal function as a predictor of patient survival. Two randomized controlled trials (RCT)(2, 3) supported this observation, demonstrating no survival benefit in patients with higher achieved Kt/v. Then guidelines were revised and a minimum Kt/v of 1,7/week was recommended but little emphasis was given to additional parameters of dialysis adequacy. As such, volume overload and sodium removal have gained major attention, since their optimization has been associated with decreased mortality in PD patients(4, 5). Inadequate sodium removal is associated with fluid overload which leads to ventricular hypertrophy and increased cardiovascular mortality(6). Individualized prescription is key for optimal sodium removal as there are differences between PD techniques (CAPD versus APD) and new strategies for sodium removal have emerged (low sodium solutions and adapted PD). In conclusion, future guidelines should address parameters associated with increased survival outcomes (sodium removal playing an important role) and abandon the current one fit all prescription model.
Collapse
|