1
|
Li Q, Chen Y, Yang M, Li P. The dietary inflammatory index (DII ®) and human papillomavirus infection: a cross-sectional study of US women. BMC Public Health 2024; 24:3031. [PMID: 39482657 PMCID: PMC11529169 DOI: 10.1186/s12889-024-20490-0] [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: 06/18/2024] [Accepted: 10/22/2024] [Indexed: 11/03/2024] Open
Abstract
BACKGROUND Diet may influence susceptibility of human papillomavirus (HPV) infection by affecting inflammation and immunity. However, the association between HPV infection and the inflammatory potential of diet has not been investigated. The research aimed to examine the correlation between HPV status and the dietary inflammatory index (DII®). METHODS We utilized data from the National Health and Nutrition Examination Survey (NHANES) 2003-2016 to investigate the correlation between DII and HPV status among 9,256 women aged 18-59 years. DII scores were calculated based on 24-hour dietary recall interviews. The association between HPV status and DII was analyzed using weighted logistic regression and restricted cubic spline (RCS). RESULTS Women with HPV infection exhibited higher DII scores than those without HPV infection. An increased likelihood of HPV infection was found to be significantly associated with higher DII scores (OR = 1.05, 95% CI: 1.01-1.09, P = 0.021), after full multivariate adjustment. Compared with the lowest tertile of DII scores, the ORs (95% CIs) for HPV infection were 1.20 (1.01, 1.42) and 1.27 (1.07, 1.51) for the second and third tertiles, respectively (P for trend = 0.006). RCS analysis showed a U-shaped relationship between DII and HPV infection, with a breakpoint identified at 0.13. CONCLUSIONS Our findings suggest that a pro-inflammatory diet is associated with an increased likelihood of HPV infection among women in the United States. Dietary interventions to reduce inflammation may help prevent HPV infection and related diseases.
Collapse
Affiliation(s)
- Qian Li
- Department of Medical Administ, Chongqing Public Health Medical Center, Chongqing, China
| | - Yu Chen
- Department of Medical Administ, Chongqing Public Health Medical Center, Chongqing, China
| | - Mei Yang
- Department of Medical Administ, Chongqing Public Health Medical Center, Chongqing, China.
| | - Peibo Li
- Department of Medical Administ, Chongqing Public Health Medical Center, Chongqing, China.
- Department of the Tuberculosis, Chongqing Public Health Medical Center, Chongqing, China.
| |
Collapse
|
2
|
Zhao H, Yang J, Wang M, Zhang H, Zhan Y, Cao Z, Gu Z, Wang Y. Effect of IL-9 neutralising antibody on pyroptosis via SGK1/NF-κB/NLRP3/GSDMD in allergic rhinitis mice. Biomed Pharmacother 2024; 177:117019. [PMID: 38917753 DOI: 10.1016/j.biopha.2024.117019] [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: 04/02/2024] [Revised: 06/10/2024] [Accepted: 06/18/2024] [Indexed: 06/27/2024] Open
Abstract
Allergic rhinitis is a common non-infectious inflammatory disease that affects approximately 15 % of people worldwide and has a complex and unclear aetiology. In recent years, pyroptosis has been found to play a role in the development of allergic rhinitis. IL-9, pyroptosis, serum and glucocorticoid-induced protein kinase 1 (SGK1), NOD-like receptor 3 (NLRP3), and nuclear factor kappa B (NF-κB) have been shown to influence each other. Herein, we aimed to explore the role of IL-9 neutralising antibody in pyroptosis involving IL-9, SGK1, NF-κB, and NLRP3 in allergic rhinitis. We observed a decrease in cytokines involved in pyroptosis and gasdermin D (GSDMD) compared with those in mice with allergic rhinitis. Further, phosphorylation of NF-κB/p65 decreased compared with that in mice with allergic rhinitis; NLRP3 and ASC also decreased, although the levels were higher than those in controls. SGK1 levels decreased compared with that in mice with allergic rhinitis and increased after using IL-9 neutralising antibodies, thus demonstrating its negative regulatory effects. The IL-9 neutralising antibody reduced the inflammatory and pyroptosis responses via SGK1 and NF-κB/NLRP3/GSDMD pathway. Our research results indicate that IL-9 regulates allergic rhinitis via the influence of SGK1 and NF-κB/NLRP3/GSDMD signalling pathway, providing new insights for developing novel drugs to treat allergic rhinitis.
Collapse
Affiliation(s)
- He Zhao
- Department of Otolaryngology Head and Neck Surgery, Shengjing Hospital of China Medical University, Shenyang 110004, PR China
| | - Jing Yang
- Department of Otolaryngology Head and Neck Surgery, Shengjing Hospital of China Medical University, Shenyang 110004, PR China
| | - Meng Wang
- Department of Otolaryngology Head and Neck Surgery, Shengjing Hospital of China Medical University, Shenyang 110004, PR China
| | - Hanxue Zhang
- Department of Laboratory, Liaoning Blood Center, Shenyang 110004, PR China
| | - Yue Zhan
- Medical Research Center, Shengjing Hospital of China Medical University, Shenyang 110004, PR China
| | - Zhiwei Cao
- Department of Otolaryngology Head and Neck Surgery, Shengjing Hospital of China Medical University, Shenyang 110004, PR China
| | - Zhaowei Gu
- Department of Otolaryngology Head and Neck Surgery, Shengjing Hospital of China Medical University, Shenyang 110004, PR China.
| | - Yunxiu Wang
- Department of Otolaryngology Head and Neck Surgery, Shengjing Hospital of China Medical University, Shenyang 110004, PR China; Department of Clinical Epidemiology, Clinical Research Center, Shengjing Hospital of China Medical University, Shenyang 110004, PR China.
| |
Collapse
|
3
|
Dong C, Lin JM, Wang Y, Zhu J, Lin L, Xu J, Du J. Exploring the Common Pathogenic Mechanisms of Psoriasis and Atopic Dermatitis: The Interaction between SGK1 and TIGIT Signaling Pathways. Inflammation 2024:10.1007/s10753-024-02115-1. [PMID: 39088121 DOI: 10.1007/s10753-024-02115-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Revised: 07/17/2024] [Accepted: 07/26/2024] [Indexed: 08/02/2024]
Abstract
This study aims to explore the common pathogenic mechanisms of psoriasis and atopic dermatitis, two T-cell-mediated autoimmune diseases. Utilizing single-cell transcriptomic sequencing data, we revealed that Treg cells primarily express TIGIT in both psoriasis and atopic dermatitis, and identified a subset of macrophages that highly express SGK1. These cells can interact with T cells via the NECTIN2-TIGIT signaling pathway, inhibiting the differentiation of T cells into a pro-inflammatory phenotype, thereby uncovering a common immunoregulatory mechanism in both diseases. Furthermore, we discovered that inhibition of SGK1 exacerbates the inflammatory response in disease models of both conditions. These findings not only provide a new perspective for a common therapeutic strategy for psoriasis and atopic dermatitis but also highlight the importance of considering these molecular interactions in future treatments. Validation of these observations through further qPCR, immunofluorescence, and animal studies has identified potential new targets for the treatment of psoriasis and atopic dermatitis.
Collapse
Affiliation(s)
- Canbin Dong
- Department of Dermatology, Shanghai Institute of Dermatology, Huashan Hospital, Fudan University, Shanghai, 200040, People's Republic of China
| | - Jui-Ming Lin
- Department of Dermatology, Shanghai Institute of Dermatology, Huashan Hospital, Fudan University, Shanghai, 200040, People's Republic of China
| | - Yilun Wang
- Department of Dermatology, Shanghai Institute of Dermatology, Huashan Hospital, Fudan University, Shanghai, 200040, People's Republic of China
| | - Junhao Zhu
- Department of Dermatology, Shanghai Institute of Dermatology, Huashan Hospital, Fudan University, Shanghai, 200040, People's Republic of China
| | - Lanmei Lin
- Department of Dermatology, Shanghai Institute of Dermatology, Huashan Hospital, Fudan University, Shanghai, 200040, People's Republic of China
| | - Jinhua Xu
- Department of Dermatology, Shanghai Institute of Dermatology, Huashan Hospital, Fudan University, Shanghai, 200040, People's Republic of China
| | - Juan Du
- Department of Dermatology, Shanghai Institute of Dermatology, Huashan Hospital, Fudan University, Shanghai, 200040, People's Republic of China.
| |
Collapse
|
4
|
Zhang Q, Tian Y, Fu Z, Wu S, Lan H, Zhou X, Shen W, Lou Y. The role of serum-glucocorticoid regulated kinase 1 in reproductive viability: implications from prenatal programming and senescence. Mol Biol Rep 2024; 51:376. [PMID: 38427115 PMCID: PMC10907440 DOI: 10.1007/s11033-024-09341-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Accepted: 02/09/2024] [Indexed: 03/02/2024]
Abstract
OBJECTIVE Organisms and cellular viability are of paramount importance to living creatures. Disruption of the balance between cell survival and apoptosis results in compromised viability and even carcinogenesis. One molecule involved in keeping this homeostasis is serum-glucocorticoid regulated kinase (SGK) 1. Emerging evidence points to a significant role of SGK1 in cell growth and survival, cell metabolism, reproduction, and life span, particularly in prenatal programming and reproductive senescence by the same token. Whether the hormone inducible SGK1 kinase is a major driver in the pathophysiological processes of prenatal programming and reproductive senescence? METHOD The PubMed/Medline, Web of Science, Embase/Ovid, and Elsevier Science Direct literature databases were searched for articles in English focusing on SGK1 published up to July 2023 RESULT: Emerging evidence is accumulating pointing to a pathophysiological role of the ubiquitously expressed SGK1 in the cellular and organismal viability. Under the regulation of specific hormones, extracellular stimuli, and various signals, SGK1 is involved in several biological processes relevant to viability, including cell proliferation and survival, cell migration and differentiation. In line, SGK1 contributes to the development of germ cells, embryos, and fetuses, whereas SGK1 inhibition leads to abnormal gametogenesis, embryo loss, and truncated reproductive lifespan. CONCLUTION SGK1 integrates a broad spectrum of effects to maintain the homeostasis of cell survival and apoptosis, conferring viability to multiple cell types as well as both simple and complex organisms, and thus ensuring appropriate prenatal development and reproductive lifespan.
Collapse
Affiliation(s)
- Qiying Zhang
- Department of Gynaecology, Hangzhou Hospital of Traditional Chinese Medicine, Hangzhou TCM Hospital Affiliated to Zhejiang Chinese Medical University, No. 453 Tiyuchang Road, Hangzhou, 310007, Zhejiang, China
| | - Ye Tian
- Department of Gynaecology, Hangzhou Hospital of Traditional Chinese Medicine, Hangzhou TCM Hospital Affiliated to Zhejiang Chinese Medical University, No. 453 Tiyuchang Road, Hangzhou, 310007, Zhejiang, China
| | - Zhujing Fu
- Jinhua Municipal Central Hospital, Jinhua, 321001, China
| | - Shuangyu Wu
- Medical School, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Huizhen Lan
- Department of Gynaecology, Hangzhou Hospital of Traditional Chinese Medicine, Hangzhou TCM Hospital Affiliated to Zhejiang Chinese Medical University, No. 453 Tiyuchang Road, Hangzhou, 310007, Zhejiang, China
| | - Xuanle Zhou
- Department of Gynaecology, Hangzhou Hospital of Traditional Chinese Medicine, Hangzhou TCM Hospital Affiliated to Zhejiang Chinese Medical University, No. 453 Tiyuchang Road, Hangzhou, 310007, Zhejiang, China
| | - Wendi Shen
- Department of Gynaecology, Hangzhou Hospital of Traditional Chinese Medicine, Hangzhou TCM Hospital Affiliated to Zhejiang Chinese Medical University, No. 453 Tiyuchang Road, Hangzhou, 310007, Zhejiang, China
| | - Yiyun Lou
- Department of Gynaecology, Hangzhou Hospital of Traditional Chinese Medicine, Hangzhou TCM Hospital Affiliated to Zhejiang Chinese Medical University, No. 453 Tiyuchang Road, Hangzhou, 310007, Zhejiang, China.
| |
Collapse
|
5
|
Qi J, Wang J, Zhang Y, Long H, Dong L, Wan P, Zuo Z, Chen W, Song Z. High-Salt-Diet (HSD) aggravates the progression of Inflammatory Bowel Disease (IBD) via regulating epithelial necroptosis. MOLECULAR BIOMEDICINE 2023; 4:28. [PMID: 37691056 PMCID: PMC10493205 DOI: 10.1186/s43556-023-00135-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Accepted: 06/14/2023] [Indexed: 09/12/2023] Open
Abstract
Due to its unclear etiology, there is no specific medicine to cure the recurrent and incurable inflammatory bowel disease (IBD). Unhealthy dietary habits unconsciously contributed to the progression of IBD, for example a High-Salt-Diet (HSD) is the most neglected and frequently adopted habit. However, the molecular mechanism of how HSD aggravates the progression of IBD has yet to remain uncovered. Herein, we focus on the hypothesis that necroptosis pathway may be involved in the process of IBD exacerbated by HSD. To this end, different gene expression (DEGs) profiles of human epithelia under hypertonic culture conditions were applied to screen candidate pathways. What's more, gene expression manipulation, immune microenvironment detection, RIPK3/MLKL gene knockout (KO), and wild-type (WT) mice were carried out to research the promotion of IBD progression under treatments of high salt intake. Based on our present results, gene expression profiles in human normal colon epithelia cell NCM460 were significantly changed under salt- or sucrose-induced hypertonic culture conditions. RIPK3 was significantly up-regulated under both conditions. Furthermore, mice colon epithelia cell CT26 growth was inhibited in a time- and dose-dependent manner by extra NaCl incubation. Autophagy, and Necroptosis pathways were activated and enhanced by LPS pretreatment. HSD significantly exacerbated DSS-induced IBD symptoms in vivo in a dose-dependent manner. Moreover, RIPK3-/- and MLKL-/- mice presented severe IBD symptoms in vivo. Overall, the results demonstrated that HSD aggravated the IBD progression via necroptosis activation, providing novel strategies and promising targets for the clinical treatment of IBD.
Collapse
Affiliation(s)
- Jialong Qi
- Department of Gastroenterology, Yunnan Digestive Endoscopy Clinical Medical Center, The First People's Hospital of Yunnan Province, Kunming, 650032, P.R. China
- Yunnan Provincial Key Laboratory of Clinical Virology, The First People's Hospital of Yunnan Province, Kunming, 650032, P.R. China
| | - Jinli Wang
- Department of Gastroenterology, Yunnan Digestive Endoscopy Clinical Medical Center, The First People's Hospital of Yunnan Province, Kunming, 650032, P.R. China
| | - Ying Zhang
- Department of Gastroenterology, Yunnan Digestive Endoscopy Clinical Medical Center, The First People's Hospital of Yunnan Province, Kunming, 650032, P.R. China
- School of Medicine, Kunming University of Science and Technology, Affiliated By The First People's Hospital of Yunnan Province, Kunming, 650504, Yunnan, P.R. China
| | - Huan Long
- Department of Gastroenterology, Yunnan Digestive Endoscopy Clinical Medical Center, The First People's Hospital of Yunnan Province, Kunming, 650032, P.R. China
- School of Medicine, Kunming University of Science and Technology, Affiliated By The First People's Hospital of Yunnan Province, Kunming, 650504, Yunnan, P.R. China
| | - Liang Dong
- Department of Gastroenterology, Yunnan Digestive Endoscopy Clinical Medical Center, The First People's Hospital of Yunnan Province, Kunming, 650032, P.R. China
- School of Medicine, Kunming University of Science and Technology, Affiliated By The First People's Hospital of Yunnan Province, Kunming, 650504, Yunnan, P.R. China
| | - Ping Wan
- Department of Gastroenterology, Yunnan Digestive Endoscopy Clinical Medical Center, The First People's Hospital of Yunnan Province, Kunming, 650032, P.R. China
- School of Medicine, Kunming University of Science and Technology, Affiliated By The First People's Hospital of Yunnan Province, Kunming, 650504, Yunnan, P.R. China
| | - Zan Zuo
- Department of Gastroenterology, Yunnan Digestive Endoscopy Clinical Medical Center, The First People's Hospital of Yunnan Province, Kunming, 650032, P.R. China.
| | - Wenjie Chen
- State Key Laboratory of Respiratory Disease, Guangdong-Hongkong-Macao Joint Laboratory of Respiratory Infectious Disease, Guangzhou Medical University, Guangzhou, 510182, P.R. China.
- Sydney Vital Translational Cancer Research Centre, Westbourne St, Sydney, NSW, 2065, Australia.
| | - Zhengji Song
- Department of Gastroenterology, Yunnan Digestive Endoscopy Clinical Medical Center, The First People's Hospital of Yunnan Province, Kunming, 650032, P.R. China.
| |
Collapse
|
6
|
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
|
7
|
Brescia C, Dattilo V, D’Antona L, Chiarella E, Tallerico R, Audia S, Rocca V, Iuliano R, Trapasso F, Perrotti N, Amato R. RANBP1, a member of the nuclear-cytoplasmic trafficking-regulator complex, is the terminal-striking point of the SGK1-dependent Th17 + pathological differentiation. Front Immunol 2023; 14:1213805. [PMID: 37441077 PMCID: PMC10333757 DOI: 10.3389/fimmu.2023.1213805] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Accepted: 06/13/2023] [Indexed: 07/15/2023] Open
Abstract
The Th17+ arrangement is critical for orchestrating both innate and acquired immune responses. In this context, the serum and glucocorticoid regulated kinase 1 (SGK1) exerts a key role in the governance of IL-23R-dependent Th17+ maturation, through the phosphorylation-dependent control of FOXO1 localization. Our previous work has shown that some of the SGK1-key functions are dependent on RAN-binding protein 1 (RANBP1), a terminal gene in the nuclear transport regulation. Here, we show that RANBP1, similarly to SGK1, is modulated during Th17+ differentiation and that RANBP1 fluctuations mediate the SGK1-dependent effects on Th17+ maturation. RANBP1, as the final effector of the SGK1 pathway, affects FOXO1 transport from the nucleus to the cytoplasm, thus enabling RORγt activation. In this light, RANBP1 represents the missing piece, in an essential and rate-limiting manner, underlying the Th17+ immune asset.
Collapse
Affiliation(s)
- Carolina Brescia
- Department of Health Science, Medical School, University “Magna Graecia” of Catanzaro, Catanzaro, Italy
- Immuno-Genetics Lab, Department of Health Science, Medical School, University “Magna Graecia” of Catanzaro, Catanzaro, Italy
| | - Vincenzo Dattilo
- Department of Experimental and Clinical Medicine, Medical School, University “Magna Graecia” of Catanzaro, Catanzaro, Italy
| | - Lucia D’Antona
- Department of Health Science, Medical School, University “Magna Graecia” of Catanzaro, Catanzaro, Italy
- Medical Genetics Unit, University Hospital, Medical School, University “Magna Graecia” of Catanzaro, Catanzaro, Italy
| | - Emanuela Chiarella
- Department of Experimental and Clinical Medicine, Medical School, University “Magna Graecia” of Catanzaro, Catanzaro, Italy
| | - Rossana Tallerico
- Microbiology and Virology Unit, “Pugliese-Ciaccio” Hospital, Catanzaro, Italy
| | - Salvatore Audia
- Department of Health Science, Medical School, University “Magna Graecia” of Catanzaro, Catanzaro, Italy
- Immuno-Genetics Lab, Department of Health Science, Medical School, University “Magna Graecia” of Catanzaro, Catanzaro, Italy
| | - Valentina Rocca
- Department of Health Science, Medical School, University “Magna Graecia” of Catanzaro, Catanzaro, Italy
- Medical Genetics Unit, University Hospital, Medical School, University “Magna Graecia” of Catanzaro, Catanzaro, Italy
| | - Rodolfo Iuliano
- Department of Health Science, Medical School, University “Magna Graecia” of Catanzaro, Catanzaro, Italy
- Medical Genetics Unit, University Hospital, Medical School, University “Magna Graecia” of Catanzaro, Catanzaro, Italy
| | - Francesco Trapasso
- Department of Experimental and Clinical Medicine, Medical School, University “Magna Graecia” of Catanzaro, Catanzaro, Italy
- Medical Genetics Unit, University Hospital, Medical School, University “Magna Graecia” of Catanzaro, Catanzaro, Italy
| | - Nicola Perrotti
- Department of Health Science, Medical School, University “Magna Graecia” of Catanzaro, Catanzaro, Italy
- Medical Genetics Unit, University Hospital, Medical School, University “Magna Graecia” of Catanzaro, Catanzaro, Italy
| | - Rosario Amato
- Department of Health Science, Medical School, University “Magna Graecia” of Catanzaro, Catanzaro, Italy
- Immuno-Genetics Lab, Department of Health Science, Medical School, University “Magna Graecia” of Catanzaro, Catanzaro, Italy
- Medical Genetics Unit, University Hospital, Medical School, University “Magna Graecia” of Catanzaro, Catanzaro, Italy
| |
Collapse
|
8
|
Abstract
Isolated systolic hypertension is associated with higher risk of cardiovascular disease and all-cause mortality. Despite being the most common form of hypertension in the elderly, it is also detectable among young and middle-aged subjects. Dietary salt (sodium chloride) intake is an important determinant of blood pressure, and high salt intake is associated with greater risk of hypertension and cardiovascular events. In most countries, habitual salt intake at all age categories largely exceeds the international recommendations. Excess salt intake, often interacting with overweight and insulin resistance, may contribute to the development and maintenance of isolated systolic hypertension in young individuals by causing endothelial dysfunction and promoting arterial stiffness through a number of mechanisms, namely increase in the renin-angiotensin-aldosterone system activity, sympathetic tone and salt-sensitivity. This short review focused on the epidemiological and clinical evidence, the mechanistic pathways and the cluster of pathophysiological factors whereby excess salt intake may favor the development and maintenance of isolated systolic hypertension in young people.
Collapse
Affiliation(s)
- Lanfranco D'Elia
- Medical School, Department of Clinical Medicine and Surgery, ESH Excellence Center of Hypertension, University of Naples Federico II, Naples, Italy
| | - Pasquale Strazzullo
- Medical School, Department of Clinical Medicine and Surgery, ESH Excellence Center of Hypertension, University of Naples Federico II, Naples, Italy -
| |
Collapse
|
9
|
Th17-Gene Expression Profile in Patients with Chronic Venous Disease and Venous Ulcers: Genetic Modulations and Preliminary Clinical Evidence. Biomolecules 2022; 12:biom12070902. [PMID: 35883458 PMCID: PMC9312858 DOI: 10.3390/biom12070902] [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: 05/25/2022] [Revised: 06/20/2022] [Accepted: 06/25/2022] [Indexed: 11/22/2022] Open
Abstract
Chronic venous disease is a condition globally widespread, resulting in a disabling pathological disorder. The CD4 + Th17+ (Cluster Differentiation 4) lymphocytes represent a regulative factor for innate immunity related to the development of complex diseases. Recently, these mechanisms have been associated with vascular disease. The aim of this work is to validate whether the Th17 response correlates with the development of CVI (Chronic venous insufficiency)and CVLUs (chronic venous limbs ulcers) and whether Th17 markers can be used, both as intrinsic risk factors and diagnostic markers, for disease development. PBL derived from peripheral blood samples of patients and controls were subjected to gene expression analysis for IL23R, IL17, SGK1, TGFβ, RORγ, FOXO1, and RANBP1 by qRT-PCR and immunoblot. A post hoc correlation, the diagnostic performance of the target genes, and multivariable analyses were properly conducted. The main expression markers of the CD4 + Th17+ switch were strongly activated in chronic venous insufficiency and in advanced ulceration. The correlation analysis demonstrated the inter-dependence on Th17’s signature modulation. ROC (Receiver Operating Characteristic) analysis defined, for the examined genes, a clinical value as the potential diagnostic markers. Multi-logistic regression studies showed that Th17 markers behave as empirical risk factors for CVD (chronic venous disease) development. Taken together, the present data provide a new hypothesis for the TH17-dependent pathogenesis of CVD, favoring the possibility for the development of new diagnostic, preventive, and therapeutic approaches.
Collapse
|
10
|
Das UN. Molecular biochemical aspects of salt (sodium chloride) in inflammation and immune response with reference to hypertension and type 2 diabetes mellitus. Lipids Health Dis 2021; 20:83. [PMID: 34334139 PMCID: PMC8327432 DOI: 10.1186/s12944-021-01507-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2021] [Accepted: 07/14/2021] [Indexed: 12/18/2022] Open
Abstract
Obesity, insulin resistance, type 2 diabetes mellitus (T2DM) and hypertension (HTN) are common that are associated with low-grade systemic inflammation. Diet, genetic factors, inflammation, and immunocytes and their cytokines play a role in their pathobiology. But the exact role of sodium, potassium, magnesium and other minerals, trace elements and vitamins in the pathogenesis of HTN and T2DM is not known. Recent studies showed that sodium and potassium can modulate oxidative stress, inflammation, alter the autonomic nervous system and induce dysfunction of the innate and adaptive immune responses in addition to their action on renin-angiotensin-aldosterone system. These actions of sodium, potassium and magnesium and other minerals, trace elements and vitamins are likely to be secondary to their action on pro-inflammatory cytokines IL-6, TNF-α and IL-17 and metabolism of essential fatty acids that may account for their involvement in the pathobiology of insulin resistance, T2DM, HTN and autoimmune diseases.
Collapse
Affiliation(s)
- Undurti N Das
- UND Life Sciences, 2221 NW 5th St, Battle Ground, WA, 98604, USA.
| |
Collapse
|
11
|
A high salt diet induces tubular damage associated with a pro-inflammatory and pro-fibrotic response in a hypertension-independent manner. Biochim Biophys Acta Mol Basis Dis 2020; 1866:165907. [DOI: 10.1016/j.bbadis.2020.165907] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2020] [Revised: 07/24/2020] [Accepted: 07/27/2020] [Indexed: 12/14/2022]
|
12
|
Liu R, Yuan M, Xu H, Chen P, Xu XS, Yang Y. Adaptive weighted sum tests via LASSO method in multi-locus family-based association analysis. Comput Biol Chem 2020; 88:107320. [PMID: 32711355 DOI: 10.1016/j.compbiolchem.2020.107320] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2019] [Revised: 03/06/2020] [Accepted: 06/22/2020] [Indexed: 01/20/2023]
Abstract
Family based multi-locus tests integrate information from individual loci by weighted averaging of the marginal statistics, and have been proven to be more efficient and robust than the single-locus tests in genetic association studies. The power depends on how much information the weights can extract from data. The currently published weighted sum methods are only applicable to either common or rare variants and may suffer from substantial power loss especially for rare variants. In this paper, we propose a novel data-driven weight to improve the power under both common and rare variant circumstances. We use the l1 regularization in Least Absolute Shrinkage and Selection Operator (LASSO) regression to construct the weight serving as a simultaneously adaptive marker selection process. Simulations for a dichotomous phenotype demonstrated that our LASSO-based approach outperformed the existing multi-locus methods in the sense of providing the highest statistical power while well controlled type I error rate under different scenarios. We also applied our methods to a real dataset for rheumatoid arthritis (GAW15 Problem 2). Two groups of alleles, in which individual SNPs had only modest and non-significant effects, were detected (P < 0.00001) using our proposed methods, whereas traditional multi-locus methods failed to identify them. In conclusion, the novel LASSO-based approach represents a superior weight-choosing strategy for multi-locus tests.
Collapse
Affiliation(s)
- Rui Liu
- Department of Statistics and Finance, University of Science and Technology of China, Hefei 230026, China
| | - Min Yuan
- School of Public Health Administration, Anhui Medical School, Hefei 230032, China
| | - Huang Xu
- Department of Statistics and Finance, University of Science and Technology of China, Hefei 230026, China
| | - Pinzhong Chen
- Jiangyin Hospital of Traditional Chinese Medicine, Wuxi 214401, China
| | - Xu Steven Xu
- Janssen Research and Development, 920 Route 202, Raritan, NJ 08869, USA.
| | - Yaning Yang
- Department of Statistics and Finance, University of Science and Technology of China, Hefei 230026, China.
| |
Collapse
|
13
|
Lee E, Kim N, Kang J, Yoon S, Lee HA, Jung H, Kim SH, Kim I. Activated pathogenic Th17 lymphocytes induce hypertension following high-fructose intake in Dahl salt-sensitive but not Dahl salt-resistant rats. Dis Model Mech 2020; 13:dmm044107. [PMID: 32179549 PMCID: PMC7272342 DOI: 10.1242/dmm.044107] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2020] [Accepted: 03/06/2020] [Indexed: 12/12/2022] Open
Abstract
High-salt intake and high-fructose intake are risk factors for hypertension via oxidative stress and inflammation. T helper (Th)17 lymphocytes play an important role in the development of hypertension. Here, we tested the hypothesis that activation of pathogenic Th17 lymphocytes induces hypertension after high-fructose intake in Dahl salt-sensitive (SS) but not Dahl salt-resistant (SR) rats. Eight-week-old male SS and SR rats were offered 20% fructose solution or tap water only for 4 weeks. Systolic blood pressure was measured by the tail-cuff method. T lymphocyte [Th17 and T regulatory (Treg)] profiling was determined via flow cytometry. The expression of Th17-related (IL-17A, IL-17RA, IL-23R and RORγt) and Treg-related (IL-10, CD25, FOXP3 and TGFβ) factors were measured via ELISA or qRT-PCR. Th17 lymphocytes isolated from high-fructose-fed SS rats were intraperitoneally injected into recipient SS and SR rats, and recombinant IL-23 protein was subcutaneously injected into SS and SR rats to induce hypertension.High-fructose intake induced hypertension via the activation of pathogenic Th17 lymphocytes in SS but not SR rats. Injection of activated Th17 lymphocytes isolated from fructose-fed SS rats induced hypertension via increase of serum IL-17A only in recipient SS rats. In addition, injection of IL-23 induced hypertension via activation of pathogenic Th17 lymphocytes only in SS rats.Thus, activation of pathogenic Th17 lymphocytes induces hypertension after high-fructose intake in SS but not SR rats. These results indicate that immunologic tolerance plays an important role in protection against hypertension in SR rats.
Collapse
Affiliation(s)
- Eunjo Lee
- Department of Pharmacology, School of Medicine, 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, Department of Biomedical Science, School of Medicine, Kyungpook National University, Daegu 41944, Republic of Korea
| | - Namkyung Kim
- Department of Pharmacology, School of Medicine, Kyungpook National University, Daegu 41944, Republic of Korea
- BK21 Plus KNU Biomedical Convergence Program, Department of Biomedical Science, School of Medicine, Kyungpook National University, Daegu 41944, Republic of Korea
| | - Jinjoo Kang
- Department of Pharmacology, School of Medicine, Kyungpook National University, Daegu 41944, Republic of Korea
- BK21 Plus KNU Biomedical Convergence Program, Department of Biomedical Science, School of Medicine, Kyungpook National University, Daegu 41944, Republic of Korea
| | - Sangwon Yoon
- Department of Pharmacology, School of Medicine, 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, Department of Biomedical Science, School of Medicine, Kyungpook National University, Daegu 41944, Republic of Korea
| | - Hae-Ahm Lee
- Medical Research Center for Bioreaction to Reactive Oxygen Species and Biomedical Science Institute, School of Medicine, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Hanna Jung
- Department of Thoracic and Cardiovascular Surgery, Kyungpook National University Hospital, Daegu 41944, Republic of Korea
| | - Sang-Hyun Kim
- Department of Pharmacology, School of Medicine, 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, Department of Biomedical Science, School of Medicine, Kyungpook National University, Daegu 41944, Republic of Korea
| | - Inkyeom Kim
- Department of Pharmacology, School of Medicine, 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, Department of Biomedical Science, School of Medicine, Kyungpook National University, Daegu 41944, Republic of Korea
| |
Collapse
|
14
|
Mattson DL. Immune mechanisms of salt-sensitive hypertension and renal end-organ damage. Nat Rev Nephrol 2019; 15:290-300. [PMID: 30804523 DOI: 10.1038/s41581-019-0121-z] [Citation(s) in RCA: 80] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Immune mechanisms have been recognized to have a role in the pathogenesis of hypertension, vascular disease and kidney damage in humans and animals for many decades. Contemporary advances in experimentation have permitted a deeper understanding of the mechanisms by which inflammation and immunity participate in cardiovascular disease, and multiple observations have demonstrated strong correlations between the discoveries made in animals and those made in patients with hypertension. Of note, striking phenotypic similarities have been observed in the infiltration of immune cells in the kidney and the development of end-organ damage in patients and animal models with sodium-sensitive hypertension. The available data suggest that an initial salt-induced increase in renal perfusion pressure, which is likely independent of immune mechanisms, induces the infiltration of immune cells into the kidney. The mechanisms mediating immune cell infiltration in the kidney are not well understood but likely involve tissue damage, the direct influence of salt to stimulate immune cell activation, sympathetic nerve stimulation or other factors. The infiltrating cells then release cytokines, free radicals and other factors that contribute to renal damage as well as increased retention of sodium and water and vascular resistance, which lead to the further development of hypertension.
Collapse
Affiliation(s)
- David L Mattson
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI, USA.
| |
Collapse
|
15
|
Abais-Battad JM, Lund H, Fehrenbach DJ, Dasinger JH, Alsheikh AJ, Mattson DL. Parental Dietary Protein Source and the Role of CMKLR1 in Determining the Severity of Dahl Salt-Sensitive Hypertension. Hypertension 2019; 73:440-448. [PMID: 30595125 DOI: 10.1161/hypertensionaha.118.11994] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Studies from our laboratory have revealed an important role for the maternal diet and the dietary protein source in the development of hypertension and renal injury in Dahl salt-sensitive (SS) rats. The current study sought to compare salt-induced hypertension, renal damage, and immune cell infiltration in the offspring of breeders fed either a casein- or gluten-based diet, with the hypothesis that offspring from gluten-fed breeders would fail to develop these SS phenotypes. When fed identical diets post-weaning, the F1 generation gluten offspring demonstrated lower mean arterial pressure (149.1±3.1 versus 162.5±5.8 mm Hg), albuminuria (166.2±34.6 versus 250.9±27.8 mg/day), and outer medullary protein casting (7.4±0.8% versus 13.1±1.3%) in response to high salt compared with the casein offspring (n=9-11). The gluten offspring also had fewer CD45+ leukocytes, CD11b/c+ monocytes/macrophages, CD3+ T cells, and CD45R+ B cells infiltrating the kidney. Analysis of the F2 generation gluten offspring also exhibited lower mean arterial pressure and renal damage compared with rats born from casein breeders (n=7-9), with no difference in renal immune cell infiltration. CMKLR1-receptor for the novel prohypertensive adipokine chemerin-was found via polymerase chain reaction array to be significantly upregulated (2.99-fold) in renal T cells isolated from F2 offspring of casein-fed versus gluten-fed parents. Furthermore, CMKLR1 inhibition via α-NETA (2-[α-naphthoyl] ethyltrimethylammonium iodide) treatment significantly attenuated renal immune cell infiltration, hypertension, and renal damage in SS rats fed high salt. Together, these data demonstrate the influence of the parental diet in determining the salt-induced hypertension, renal damage, and inflammatory phenotype of the offspring.
Collapse
Affiliation(s)
| | - Hayley Lund
- From the Department of Physiology, Medical College of Wisconsin, Milwaukee
| | | | | | - Ammar J Alsheikh
- From the Department of Physiology, Medical College of Wisconsin, Milwaukee
| | - David L Mattson
- From the Department of Physiology, Medical College of Wisconsin, Milwaukee
| |
Collapse
|
16
|
Fucà G, Galli G, Poggi M, Lo Russo G, Proto C, Imbimbo M, Vitali M, Ganzinelli M, Lanti C, Molino G, Stangoni F, Zilembo N, de Braud F, Garassino MC, Signorelli D. Low Baseline Serum Sodium Concentration Is Associated with Poor Clinical Outcomes in Metastatic Non-Small Cell Lung Cancer Patients Treated with Immunotherapy. Target Oncol 2019; 13:795-800. [PMID: 30306460 DOI: 10.1007/s11523-018-0599-5] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
BACKGROUND A consistent percentage of patients with metastatic non-small cell lung cancer (NSCLC) derives no or only marginal benefit from immunotherapy (IO). OBJECTIVE Since serum sodium has been linked to both prognosis in NSCLC and modulation of immune cells activity, we aimed to assess the association between low baseline serum sodium concentration (≤ 135 mEq/L) and clinical outcomes of patients with metastatic NSCLC treated with IO. PATIENTS AND METHODS We included metastatic NSCLC patients treated with checkpoint inhibitors in our department from April 2013 to April 2018 with available baseline serum sodium concentration. Demographics, clinical and pathological characteristics were collected. Survival analyses were performed using the Kaplan-Meier method and the Cox proportional-hazards model. RESULTS Of 197 patients included, 26 (13%) presented low baseline serum sodium concentration. Patients in the low sodium cohort experienced a poorer disease control rate (OR 0.36; 95% CI, 0.15-0.86; Wald test P = .02), median overall survival (OS) (2.8 vs. 11.6 months; HR 3.00; 95% CI, 1.80-4.80; P < .001) and progression-free survival (PFS) (1.8 vs. 3.3 months; HR 2.60; 95% CI, 1.70-3.90; P < .001) compared to patients in the control cohort. At multivariate analyses, low baseline serum sodium concentration was independently associated with disease control and OS, but not with PFS. CONCLUSIONS Our study showed for the first time that low baseline serum sodium concentration is associated with impaired clinical outcomes in patients with metastatic NSCLC treated with IO. The role of serum sodium concentration in this setting warrants further pre-clinical and clinical investigation.
Collapse
Affiliation(s)
- Giovanni Fucà
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, Via G. Venezian, 1, 20133, Milan, Italy.
| | - Giulia Galli
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, Via G. Venezian, 1, 20133, Milan, Italy
| | - Marta Poggi
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, Via G. Venezian, 1, 20133, Milan, Italy
| | - Giuseppe Lo Russo
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, Via G. Venezian, 1, 20133, Milan, Italy
| | - Claudia Proto
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, Via G. Venezian, 1, 20133, Milan, Italy
| | - Martina Imbimbo
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, Via G. Venezian, 1, 20133, Milan, Italy
| | - Milena Vitali
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, Via G. Venezian, 1, 20133, Milan, Italy
| | - Monica Ganzinelli
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, Via G. Venezian, 1, 20133, Milan, Italy
| | - Claudia Lanti
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, Via G. Venezian, 1, 20133, Milan, Italy
| | - Giuliano Molino
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, Via G. Venezian, 1, 20133, Milan, Italy
| | - Fabiano Stangoni
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, Via G. Venezian, 1, 20133, Milan, Italy
| | - Nicoletta Zilembo
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, Via G. Venezian, 1, 20133, Milan, Italy
| | - Filippo de Braud
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, Via G. Venezian, 1, 20133, Milan, Italy.,Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| | - Marina Chiara Garassino
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, Via G. Venezian, 1, 20133, Milan, Italy
| | - Diego Signorelli
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, Via G. Venezian, 1, 20133, Milan, Italy
| |
Collapse
|
17
|
Abais-Battad JM, Alsheikh AJ, Pan X, Fehrenbach DJ, Dasinger JH, Lund H, Roberts ML, Kriegel AJ, Cowley AW, Kidambi S, Kotchen TA, Liu P, Liang M, Mattson DL. Dietary Effects on Dahl Salt-Sensitive Hypertension, Renal Damage, and the T Lymphocyte Transcriptome. Hypertension 2019; 74:854-863. [PMID: 31476910 DOI: 10.1161/hypertensionaha.119.12927] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The Dahl salt-sensitive (SS) rat is an established model of SS hypertension and renal damage. In addition to salt, other dietary components were shown to be important determinants of hypertension in SS rats. With previous work eliminating the involvement of genetic differences, grain-fed SS rats from Charles River Laboratories (SS/CRL; 5L2F/5L79) were less susceptible to salt-induced hypertension and renal damage compared with purified diet-fed SS rats bred at the Medical College of Wisconsin (SS/MCW; 0.4% NaCl, AIN-76A). With the known role of immunity in hypertension, the present study characterized the immune cells infiltrating SS/MCW and SS/CRL kidneys via flow cytometry and RNA sequencing in T-cells isolated from the blood and kidneys of rats maintained on their respective parental diet or on 3 weeks of high salt (4.0% NaCl, AIN-76A). SS/CRL rats were protected from salt-induced hypertension (116.5±1.2 versus 141.9±14.4 mm Hg), albuminuria (21.7±3.5 versus 162.9±22.2 mg/d), and renal immune cell infiltration compared with SS/MCW. RNA-seq revealed >50% of all annotated genes in the entire transcriptome to be significantly differentially expressed in T-cells isolated from blood versus kidney, regardless of colony or chow. Pathway analysis of significantly differentially expressed genes between low and high salt conditions demonstrated changes related to inflammation in SS/MCW renal T-cells compared with metabolism-related pathways in SS/CRL renal T-cells. These functional and transcriptomic T-cell differences between SS/MCW and SS/CRL show that dietary components in addition to salt may influence immunity and the infiltration of immune cells into the kidney, ultimately impacting susceptibility to salt-induced hypertension and renal damage.
Collapse
Affiliation(s)
- Justine M Abais-Battad
- From the Department of Physiology (J.M.A.-B., A.J.A., X.P., D.J.F., J.H.D., H.L., M.L.R., A.J.K., A.W.C., P.L., M.L., D.L.M.), Medical College of Wisconsin
| | - Ammar J Alsheikh
- From the Department of Physiology (J.M.A.-B., A.J.A., X.P., D.J.F., J.H.D., H.L., M.L.R., A.J.K., A.W.C., P.L., M.L., D.L.M.), Medical College of Wisconsin
| | - Xiaoqing Pan
- From the Department of Physiology (J.M.A.-B., A.J.A., X.P., D.J.F., J.H.D., H.L., M.L.R., A.J.K., A.W.C., P.L., M.L., D.L.M.), Medical College of Wisconsin
| | - Daniel J Fehrenbach
- From the Department of Physiology (J.M.A.-B., A.J.A., X.P., D.J.F., J.H.D., H.L., M.L.R., A.J.K., A.W.C., P.L., M.L., D.L.M.), Medical College of Wisconsin
| | - John Henry Dasinger
- From the Department of Physiology (J.M.A.-B., A.J.A., X.P., D.J.F., J.H.D., H.L., M.L.R., A.J.K., A.W.C., P.L., M.L., D.L.M.), Medical College of Wisconsin
| | - Hayley Lund
- From the Department of Physiology (J.M.A.-B., A.J.A., X.P., D.J.F., J.H.D., H.L., M.L.R., A.J.K., A.W.C., P.L., M.L., D.L.M.), Medical College of Wisconsin
| | - Michelle L Roberts
- From the Department of Physiology (J.M.A.-B., A.J.A., X.P., D.J.F., J.H.D., H.L., M.L.R., A.J.K., A.W.C., P.L., M.L., D.L.M.), Medical College of Wisconsin
| | - Alison J Kriegel
- From the Department of Physiology (J.M.A.-B., A.J.A., X.P., D.J.F., J.H.D., H.L., M.L.R., A.J.K., A.W.C., P.L., M.L., D.L.M.), Medical College of Wisconsin
| | - Allen W Cowley
- From the Department of Physiology (J.M.A.-B., A.J.A., X.P., D.J.F., J.H.D., H.L., M.L.R., A.J.K., A.W.C., P.L., M.L., D.L.M.), Medical College of Wisconsin
| | - Srividya Kidambi
- Department of Medicine (S.K., T.A.K.), Medical College of Wisconsin
| | | | - Pengyuan Liu
- From the Department of Physiology (J.M.A.-B., A.J.A., X.P., D.J.F., J.H.D., H.L., M.L.R., A.J.K., A.W.C., P.L., M.L., D.L.M.), Medical College of Wisconsin
| | - Mingyu Liang
- From the Department of Physiology (J.M.A.-B., A.J.A., X.P., D.J.F., J.H.D., H.L., M.L.R., A.J.K., A.W.C., P.L., M.L., D.L.M.), Medical College of Wisconsin
| | - David L Mattson
- From the Department of Physiology (J.M.A.-B., A.J.A., X.P., D.J.F., J.H.D., H.L., M.L.R., A.J.K., A.W.C., P.L., M.L., D.L.M.), Medical College of Wisconsin
| |
Collapse
|
18
|
Willebrand R, Hamad I, Van Zeebroeck L, Kiss M, Bruderek K, Geuzens A, Swinnen D, Côrte-Real BF, Markó L, Lebegge E, Laoui D, Kemna J, Kammertoens T, Brandau S, Van Ginderachter JA, Kleinewietfeld M. High Salt Inhibits Tumor Growth by Enhancing Anti-tumor Immunity. Front Immunol 2019; 10:1141. [PMID: 31214164 PMCID: PMC6557976 DOI: 10.3389/fimmu.2019.01141] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2019] [Accepted: 05/07/2019] [Indexed: 02/02/2023] Open
Abstract
Excess salt intake could affect the immune system by shifting the immune cell balance toward a pro-inflammatory state. Since this shift of the immune balance is thought to be beneficial in anti-cancer immunity, we tested the impact of high salt diets on tumor growth in mice. Here we show that high salt significantly inhibited tumor growth in two independent murine tumor transplantation models. Although high salt fed tumor-bearing mice showed alterations in T cell populations, the effect seemed to be largely independent of adaptive immune cells. In contrast, depletion of myeloid-derived suppressor cells (MDSCs) significantly reverted the inhibitory effect on tumor growth. In line with this, high salt conditions almost completely blocked murine MDSC function in vitro. Importantly, similar effects were observed in human MDSCs isolated from cancer patients. Thus, high salt conditions seem to inhibit tumor growth by enabling more pronounced anti-tumor immunity through the functional modulation of MDSCs. Our findings might have critical relevance for cancer immunotherapy.
Collapse
Affiliation(s)
- Ralf Willebrand
- VIB Laboratory of Translational Immunomodulation, VIB Center for Inflammation Research, University of Hasselt, Campus Diepenbeek, Hasselt, Belgium
| | - Ibrahim Hamad
- VIB Laboratory of Translational Immunomodulation, VIB Center for Inflammation Research, University of Hasselt, Campus Diepenbeek, Hasselt, Belgium
| | - Lauren Van Zeebroeck
- VIB Laboratory of Translational Immunomodulation, VIB Center for Inflammation Research, University of Hasselt, Campus Diepenbeek, Hasselt, Belgium
| | - Máté Kiss
- Cellular and Molecular Immunology Lab, Vrije Universiteit Brussel, Brussels, Belgium.,Myeloid Cell Immunology Lab, VIB Center for Inflammation Research, Brussels, Belgium
| | - Kirsten Bruderek
- Research Division, Department of Otorhinolaryngology, West German Cancer Center, University Hospital Essen, Essen, Germany
| | - Anneleen Geuzens
- VIB Laboratory of Translational Immunomodulation, VIB Center for Inflammation Research, University of Hasselt, Campus Diepenbeek, Hasselt, Belgium
| | - Dries Swinnen
- VIB Laboratory of Translational Immunomodulation, VIB Center for Inflammation Research, University of Hasselt, Campus Diepenbeek, Hasselt, Belgium
| | - Beatriz Fernandes Côrte-Real
- VIB Laboratory of Translational Immunomodulation, VIB Center for Inflammation Research, University of Hasselt, Campus Diepenbeek, Hasselt, Belgium
| | - Lajos Markó
- Experimental and Clinical Research Center, A Joint Cooperation of Max Delbrück Center for Molecular Medicine and Charité University Medicine Berlin, Berlin, Germany
| | - Els Lebegge
- Cellular and Molecular Immunology Lab, Vrije Universiteit Brussel, Brussels, Belgium.,Myeloid Cell Immunology Lab, VIB Center for Inflammation Research, Brussels, Belgium
| | - Damya Laoui
- Cellular and Molecular Immunology Lab, Vrije Universiteit Brussel, Brussels, Belgium.,Myeloid Cell Immunology Lab, VIB Center for Inflammation Research, Brussels, Belgium
| | - Josephine Kemna
- Institute of Immunology, Charité - Universitätsmedizin Berlin, Berlin, Germany.,Max Delbrück Center for Molecular Medicine, Berlin, Germany
| | - Thomas Kammertoens
- Institute of Immunology, Charité - Universitätsmedizin Berlin, Berlin, Germany.,Max Delbrück Center for Molecular Medicine, Berlin, Germany
| | - Sven Brandau
- Research Division, Department of Otorhinolaryngology, West German Cancer Center, University Hospital Essen, Essen, Germany
| | - Jo A Van Ginderachter
- Cellular and Molecular Immunology Lab, Vrije Universiteit Brussel, Brussels, Belgium.,Myeloid Cell Immunology Lab, VIB Center for Inflammation Research, Brussels, Belgium
| | - Markus Kleinewietfeld
- VIB Laboratory of Translational Immunomodulation, VIB Center for Inflammation Research, University of Hasselt, Campus Diepenbeek, Hasselt, Belgium
| |
Collapse
|
19
|
Du YN, Tang XF, Xu L, Chen WD, Gao PJ, Han WQ. SGK1-FoxO1 Signaling Pathway Mediates Th17/Treg Imbalance and Target Organ Inflammation in Angiotensin II-Induced Hypertension. Front Physiol 2018; 9:1581. [PMID: 30524295 PMCID: PMC6262360 DOI: 10.3389/fphys.2018.01581] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2018] [Accepted: 10/23/2018] [Indexed: 01/17/2023] Open
Abstract
It has been demonstrated that serum/glucocorticoid regulated kinase 1 (SGK1) and the downstream transcription factor forkhead box O1 (FoxO1) plays a critical role in the differentiation of T helper 17 cells/regulatory T cells (Th17/Treg). In the present study, we hypothesized that this SGK1-FoxO1 signaling pathway is involved in Th17/Treg imbalance and target organ damage in angiotensin II (AngII)-induced hypertensive mice. Results show that SGK1 inhibitor EMD638683 significantly reversed renal dysfunction and cardiac dysfunction in echocardiography as indicated by decreased blood urine nitrogen and serum creatinine in AngII-infused mice. Flow cytometric assay shows that there was significant Th17/Treg imbalance in spleen and in renal/cardiac infiltrating lymphocytes as indicated by the increased Th17 cells (CD4+-IL17A+ cells) and decreased Treg cells (CD4+-Foxp3+). Consistently, real-time PCR shows that Th17-related cytokines including IL-17A, IL-23, and tumor necrosis factor α (TNF-α) was increased and Treg-related cytokine IL-10 was decreased in renal and cardiac infiltrating lymphocytes in AngII-infused mice. Meanwhile, SGK1 protein level, as well as its phosphorylation and activity, was significantly increased in spleen in AngII-infused rats. Furthermore, it was found that splenic phosphorylated FoxO1 was significantly increased, whereas total FoxO1 in nuclear preparation was significantly decreased in AngII-infused mice, suggesting that increased FoxO1 phosphorylation initiate its translocation from cytoplasm to nucleus. Notably, all changes were significantly inhibited by the treatment of EMD638683. These results suggest that SGK1 was involved in Th17/Treg imbalance and target organ damage in AngII-induced hypertension.
Collapse
Affiliation(s)
- Ya-Nan Du
- Shanghai Key Laboratory of Hypertension, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Shanghai Institute of Hypertension, Shanghai, China
| | - Xiao-Feng Tang
- Shanghai Key Laboratory of Hypertension, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Shanghai Institute of Hypertension, Shanghai, China
| | - Lian Xu
- Shanghai Key Laboratory of Hypertension, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Laboratory of Vascular Biology, Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Wen-Dong Chen
- Shanghai Key Laboratory of Hypertension, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Shanghai Institute of Hypertension, Shanghai, China
| | - Ping-Jin Gao
- Shanghai Key Laboratory of Hypertension, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Shanghai Institute of Hypertension, Shanghai, China.,Laboratory of Vascular Biology, Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Wei-Qing Han
- Shanghai Key Laboratory of Hypertension, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Shanghai Institute of Hypertension, Shanghai, China.,Laboratory of Vascular Biology, Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| |
Collapse
|
20
|
Abais-Battad JM, Mattson DL. Influence of dietary protein on Dahl salt-sensitive hypertension: a potential role for gut microbiota. Am J Physiol Regul Integr Comp Physiol 2018; 315:R907-R914. [PMID: 30133303 PMCID: PMC6295491 DOI: 10.1152/ajpregu.00399.2017] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2017] [Revised: 07/30/2018] [Accepted: 08/17/2018] [Indexed: 02/07/2023]
Abstract
High blood pressure affects 1.39 billion adults across the globe and is the leading preventable cause of death worldwide. Hypertension is a multifaceted disease with known genetic and environmental factors contributing to its progression. Our studies utilizing the Dahl salt-sensitive (SS) rat have demonstrated the remarkable influence of dietary protein and maternal environment on the development of hypertension and renal damage in response to high salt. There is growing interest in the relationship between the microbiome and hypertension, with gut dysbiosis being correlated to a number of pathologies. This review summarizes the current literature regarding the interplay among dietary protein, the gut microbiota, and hypertension. These studies may provide insight into the effects we have observed between diet and hypertension in Dahl SS rats and, we hope, lead to new perspectives where potential dietary interventions or microbiota manipulations could serve as plausible therapies for hypertension.
Collapse
Affiliation(s)
| | - David L Mattson
- Department of Physiology, Medical College of Wisconsin , Milwaukee, Wisconsin
| |
Collapse
|
21
|
The role of dietary sodium in autoimmune diseases: The salty truth. Autoimmun Rev 2018; 17:1069-1073. [PMID: 30213699 DOI: 10.1016/j.autrev.2018.05.007] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2018] [Accepted: 05/08/2018] [Indexed: 02/06/2023]
Abstract
Autoimmune diseases are a group of heterogeneous condition that occur secondary to the intrinsic loss of tolerance to self- antigens. In genetically susceptible individuals, the complex interplay of environmental factors and epigenetic deregulations have been proposed to drive disease etiopathogenesis. Various environmental variables have been identified including viral infections, exposure to pollutants, stress and dietary factors. Sodium, a major constituent of salt is essential for mammalian physiology. However, high salt intake may play a role in the development of autoimmune diseases. Several lines of evidence point toward the role of high sodium intake in reversing the suppressive effects of Regulatory T cells (Tregs) and instead promoting cellular shift toward T-helper (Th)-1 and Th17 pro-inflammatory phenotypes. These effects have been attributed to cascade of events that ultimately results in downstream activation of serum glucocorticoid kinase 1 (Sgk1). In vivo, various autoimmune animal models have confirmed the role of high sodium diet in the emergence and the exacerbation of autoimmune conditions including for instance Experimental Autoimmune Encephalomyelitis model for multiple sclerosis, MRL/lpr mouse model for lupus nephritis, collagen induced arthritis model for rheumatoid arthritis, and dextran sulfate sodium induced colitis, and TNBS-induced colitis models for Crohn's disease. Clinical epidemiological studies are scarce. High sodium intake was associated with increased risk of rheumatoid arthritis disease emergence. In multiple sclerosis, some studies suggest a relation to clinical exacerbation rates however other studies did not corroborate these results. Taken together, high dietary salt intake plays a role in the spectrum of autoimmune disease etiology. Further research is warranted to better characterize such relationship and assist in identifying individuals that would benefit from dietary salt restriction.
Collapse
|
22
|
Spagnuolo R, Dattilo V, D'Antona L, Cosco C, Tallerico R, Ventura V, Conforti F, Camastra C, Mancina RM, Catalogna G, Cosco V, Iuliano R, Carbone E, Perrotti N, Amato R, Doldo P. Deregulation of SGK1 in Ulcerative Colitis: A Paradoxical Relationship Between Immune Cells and Colonic Epithelial Cells. Inflamm Bowel Dis 2018; 24:1967-1977. [PMID: 29788407 DOI: 10.1093/ibd/izy158] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/19/2017] [Indexed: 12/11/2022]
Abstract
BACKGROUND Inflammatory bowel disease (IBD) is due to the interaction of genetic and environmental factors that trigger an unbalanced immune response ultimately resulting in the peculiar inflammatory reaction. Experimental models of IBD point to a role of T-cell-derived cytokines (Th17) and to SGK1 as mediator of the Th17 switch. We hypothesize that SGK1, a salt inducible kinase, directs lymphocytic behavior and tissue damage. METHODS Eleven controls and 32 ulcerative colitis (UC) patients were randomized according to endoscopic Mayo score. Mucosal biopsies from different intestinal tracts were analyzed by immunohistochemistry and quantitative real-time polymerase chain reaction to check the expression of disease markers including SGK1. Peripheral blood mononuclear cells (PBMCs) from patients and controls were analyzed by fluorescence-activated cell sorting. Finally, an in vitro cell model was developed to test the hypothesis. RESULTS SGK1 mRNA and protein expression in lesional areas of UC patients were lower than in normal peri-lesional areas of the same patients and in normal tissues of healthy controls. SGK1 expression was increased in PBMCs from UC patients, particularly in the CD4+ cell population, enriched in Th17 cells. IL17/IL13 was increased in patients and correlated with SGK1 expression. Genetically engineered Jurkat cells confirmed the effect of SGK1 overexpression on viability of RKO cells. CONCLUSIONS These observations suggest a pathogenic mechanism whereby SGK1 overexpression in CD4+ T cells induces the secretion of the inflammatory cytokines IL17 and IL13, which downregulate the expression of SGK1 in target tissues. Our data suggest a novel hypothesis in the pathogenesis of UC, integrating colonic epithelial cells and lymphocytes.
Collapse
Affiliation(s)
- Rocco Spagnuolo
- Departments of "Scienze Mediche e Chirurgiche,", Catanzaro, Italy
| | | | - Lucia D'Antona
- Departments of "Scienze della Salute,", Catanzaro, Italy
| | - Cristina Cosco
- Departments of "Scienze Mediche e Chirurgiche,", Catanzaro, Italy
| | - Rossana Tallerico
- Departments of "Medicina Sperimentale e Clinica," University "Magna Graecia" of Catanzaro, Catanzaro, Italy
| | - Valeria Ventura
- Departments of "Medicina Sperimentale e Clinica," University "Magna Graecia" of Catanzaro, Catanzaro, Italy
| | | | | | - Rosellina M Mancina
- Department of Molecular and Clinical Medicine, University of Gothenburg, Gothenburg, Sweden
| | | | - Vincenzo Cosco
- Departments of "Scienze Mediche e Chirurgiche,", Catanzaro, Italy
| | | | - Ennio Carbone
- Departments of "Medicina Sperimentale e Clinica," University "Magna Graecia" of Catanzaro, Catanzaro, Italy.,Department of Microbiology Cell and Tumor Biology (MTC), Karolinska Institutet, Stockholm, Sweden
| | | | - Rosario Amato
- Departments of "Scienze della Salute,", Catanzaro, Italy
| | - Patrizia Doldo
- Departments of "Scienze Mediche e Chirurgiche,", Catanzaro, Italy
| |
Collapse
|
23
|
Wenzel UO, Bode M, Kurts C, Ehmke H. Salt, inflammation, IL-17 and hypertension. Br J Pharmacol 2018; 176:1853-1863. [PMID: 29767465 DOI: 10.1111/bph.14359] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2018] [Revised: 04/08/2018] [Accepted: 04/17/2018] [Indexed: 12/18/2022] Open
Abstract
Traditionally, arterial hypertension and subsequent end-organ damage have been attributed to haemodynamic factors, but increasing evidence indicates that inflammation also contributes to the deleterious consequences of this disease. The immune system has evolved to prevent invasion of foreign microorganisms and to promote tissue healing after injury. However, this beneficial activity comes at a cost of collateral damage when the immune system overreacts to internal injury, such as prehypertension. Over the past few years, important findings have revolutionized hypertension research. Firstly, in 2007, a seminal paper showed that adaptive immunity is involved in the pathogenesis of hypertension. Secondly, salt storage in the skin and its consequences for cardiovascular physiology were discovered. Thirdly, after the discovery that salt promotes the differentiation of CD4+ T cells into TH 17 cells, it was demonstrated that salt directly changes several cells of the innate and adaptive immune system and aggravates autoimmune disease but may improve antimicrobial defence. Herein, we will review pathways of activation of immune cells by salt in hypertension as the framework for understanding the multiple roles of salt and immunity in arterial hypertension and autoimmune disease. LINKED ARTICLES: This article is part of a themed section on Immune Targets in Hypertension. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v176.12/issuetoc.
Collapse
Affiliation(s)
- Ulrich O Wenzel
- III. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Marlies Bode
- III. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Christian Kurts
- Institute of Molecular Medicine and Experimental Immunology, Rheinische Friedrich-Wilhelms University, Bonn, Germany
| | - Heimo Ehmke
- Department of Cellular and Integrative Physiology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| |
Collapse
|
24
|
Toussirot E, Béreau M, Vauchy C, Saas P. Could Sodium Chloride be an Environmental Trigger for Immune-Mediated Diseases? An Overview of the Experimental and Clinical Evidence. Front Physiol 2018; 9:440. [PMID: 29740348 PMCID: PMC5928237 DOI: 10.3389/fphys.2018.00440] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2017] [Accepted: 04/06/2018] [Indexed: 12/31/2022] Open
Abstract
Immune mediated diseases (IMDs) are complex chronic inflammatory diseases involving genetic and environmental factors. Salt intake has been proposed as a diet factor that can influence the immune response. Indeed, experimental data report the influence of sodium chloride on the differentiation of naive CD4+ T cells into IL-17 secreting T helper (Th) cells (Th17 cells), by a mechanism involving the serum glucocorticoid kinase-1 (SGK1) that promotes the expression of the IL-23 receptor (IL-23R). The IL-23/IL-23R is critical for pathogenic inflammatory Th17 cell differentiation. Experimental data in murine models of arthritis, colitis and encephalomyelitis corroborate these findings. This manuscript reviews the current knowledge on the effects of sodium chloride on innate and adaptive immunity. We also performed a systematic literature review for clinical studies examining the relationships between salt consumption and the development or the activity/severity of the most common IMDs mediated by the IL-23/Th17 pathway, i.e., rheumatoid arthritis (RA), multiple sclerosis (MS), and Crohn's disease (CD). Nine studies were found, 4 in RA, 4 in MS and 1 in CD. An association was found between developments of anti-citrullinated protein antibody (ACPA) positive RA in smokers and salt intake, but these results were not confirmed in another study. For MS, no association was observed in pediatric subjects while in adult patients, a link was found between salt intake and disease activity. However, this result was not confirmed in another study. These conflicting results highlight the fact that further evaluation in human IMDs is required. Moreover, physicians need to develop clinical trials with diet interventions to evaluate the impact of low salt intake on disease activity/severity of IMDs.
Collapse
Affiliation(s)
- Eric Toussirot
- Institut National de la Santé et de la Recherche Médicale CIC-1431, Centre d'Investigation Clinique Biothérapie, University Hospital Besançon, Besançon, France.,Fédération Hospitalo-Universitaire INCREASE, University Hospital Besançon, Besançon, France.,Rhumatologie, University Hospital Besançon, Besançon, France.,Département de Thérapeutique et EPILAB EA4266: "Epigénétique des Infections Virales et des Maladies Inflammatoires", Université Bourgogne Franche-Comté, Besançon, France
| | - Matthieu Béreau
- Department of Neurology, University Hospital Besançon, Besançon, France
| | - Charline Vauchy
- Institut National de la Santé et de la Recherche Médicale CIC-1431, Centre d'Investigation Clinique Biothérapie, University Hospital Besançon, Besançon, France.,Fédération Hospitalo-Universitaire INCREASE, University Hospital Besançon, Besançon, France
| | - Philippe Saas
- Institut National de la Santé et de la Recherche Médicale CIC-1431, Centre d'Investigation Clinique Biothérapie, University Hospital Besançon, Besançon, France.,Fédération Hospitalo-Universitaire INCREASE, University Hospital Besançon, Besançon, France.,Institut National de la Santé et de la Recherche Médicale U1098, Etablissement Français du Sang Bourgogne Franche-Comté, Université Bourgogne Franche-Comté, Interactions Hôte-Greffon-Tumeurs, LabEx LipSTIC, Besançon, France
| |
Collapse
|
25
|
Potential Interplay between Hyperosmolarity and Inflammation on Retinal Pigmented Epithelium in Pathogenesis of Diabetic Retinopathy. Int J Mol Sci 2018; 19:ijms19041056. [PMID: 29614818 PMCID: PMC5979527 DOI: 10.3390/ijms19041056] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2018] [Revised: 03/21/2018] [Accepted: 03/30/2018] [Indexed: 12/15/2022] Open
Abstract
Diabetic retinopathy is a frequent eyesight threatening complication of type 1 and type 2 diabetes. Under physiological conditions, the inner and the outer blood-retinal barriers protect the retina by regulating ion, protein, and water flux into and out of the retina. During diabetic retinopathy, many factors, including inflammation, contribute to the rupture of the inner and/or the outer blood-retinal barrier. This rupture leads the development of macular edema, a foremost cause of sight loss among diabetic patients. Under these conditions, it has been speculated that retinal pigmented epithelial cells, that constitute the outer blood-retinal barrier, may be subjected to hyperosmolar stress resulting from different mechanisms. Herein, we review the possible origins and consequences of hyperosmolar stress on retinal pigmented epithelial cells during diabetic retinopathy, with a special focus on the intimate interplay between inflammation and hyperosmolar stress, as well as the current and forthcoming new pharmacotherapies for the treatment of such condition.
Collapse
|
26
|
Hu Y, Sparks JA, Malspeis S, Costenbader KH, Hu FB, Karlson EW, Lu B. Long-term dietary quality and risk of developing rheumatoid arthritis in women. Ann Rheum Dis 2017; 76:1357-1364. [PMID: 28137914 PMCID: PMC5556680 DOI: 10.1136/annrheumdis-2016-210431] [Citation(s) in RCA: 92] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2016] [Revised: 12/04/2016] [Accepted: 01/08/2017] [Indexed: 01/03/2023]
Abstract
OBJECTIVES To evaluate the association between long-term dietary quality, measured by the 2010 Alternative Healthy Eating Index, and risk of rheumatoid arthritis (RA) in women. METHODS We prospectively followed 76 597 women in the Nurses' Health Study aged 30-55 years and 93 392 women in the Nurses' Health Study II aged 25-42 years at baseline and free from RA or other connective tissue diseases. The lifestyle, environmental exposure and anthropometric information were collected at baseline and updated biennially. Cumulative follow-up rates were more than 90% for both cohorts. The primary outcome was RA alone with two subtypes of the disease: seropositive and seronegative RA. RESULTS During 3 678 104 person-years, 1007 RA cases were confirmed. In the multivariable-adjusted model, long-term adherence to healthy eating patterns was marginally associated with reduced RA risk. To assess potential effect modification by age at diagnosis, we stratified by age. Among women aged ≤55 years, better quality diet was associated with lower RA risk (HRQ4 vs Q1: 0.67; 95% CI 0.51 to 0.88; p trend: 0.002), but no significant association was found for women aged >55 years (p interaction: 0.005). When stratifying by serostatus, the inverse association among those aged ≤55 years was strongest for seropositive RA (HRQ4 vs Q1: 0.60; 95% CI 0.42 to 0.86; p trend: 0.003). CONCLUSIONS A healthier diet was associated with a reduced risk of RA occurring at 55 years of age or younger, particularly seropositive RA.
Collapse
Affiliation(s)
- Yang Hu
- Departments of Nutrition, Harvard School of Public Health, Boston, MA, USA
| | - Jeffrey A. Sparks
- Division of Rheumatology, Immunology and Allergy, Brigham & Women’s Hospital and Harvard Medical School, Boston, MA, USA
| | - Susan Malspeis
- Division of Rheumatology, Immunology and Allergy, Brigham & Women’s Hospital and Harvard Medical School, Boston, MA, USA
| | - Karen H. Costenbader
- Division of Rheumatology, Immunology and Allergy, Brigham & Women’s Hospital and Harvard Medical School, Boston, MA, USA
| | - Frank B. Hu
- Departments of Nutrition and Epidemiology, Harvard School of Public Health, USA
- Channing Division of Network Medicine, Brigham and Women’s Hospital and Harvard Medical School (FBH), Boston, MA, USA
| | - Elizabeth W. Karlson
- Division of Rheumatology, Immunology and Allergy, Brigham & Women’s Hospital and Harvard Medical School, Boston, MA, USA
| | - Bing Lu
- Division of Rheumatology, Immunology and Allergy, Brigham & Women’s Hospital and Harvard Medical School, Boston, MA, USA
| |
Collapse
|
27
|
Dahan S, Segal Y, Shoenfeld Y. Dietary factors in rheumatic autoimmune diseases: a recipe for therapy? Nat Rev Rheumatol 2017; 13:348-358. [PMID: 28405001 DOI: 10.1038/nrrheum.2017.42] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Today, we are facing a new era of digitization in the health care system, and with increased access to health care information has come a growing demand for safe, cost-effective and easy to administer therapies. Dietary habits have a crucial influence on human health, affecting an individual's risk for hypertension, heart disease and stroke, as well as influencing the risk of developing of cancer. Moreover, an individual's lifestyle choices can greatly influence the progression and manifestation of chronic autoimmune rheumatic diseases. In light of these effects, it makes sense that the search for additional therapies to attenuate such diseases would include investigations into lifestyle modifications. When considering the complex web of factors that influence autoimmunity, it is not surprising to find that several dietary elements are involved in disease progression or prevention. In this Review, several common nutritional components of the human diet are presented, and the evidence for their effects on rheumatic diseases is discussed.
Collapse
Affiliation(s)
- Shani Dahan
- The Zabludowicz Centre for Autoimmune Diseases, Chaim Sheba Medical Centre, Derech Sheba 2, Tel-Hashomer, Ramat-Gan 52621, Israel
| | - Yahel Segal
- The Zabludowicz Centre for Autoimmune Diseases, Chaim Sheba Medical Centre, Derech Sheba 2, Tel-Hashomer, Ramat-Gan 52621, Israel
| | - Yehuda Shoenfeld
- The Zabludowicz Centre for Autoimmune Diseases, Chaim Sheba Medical Centre, Derech Sheba 2, Tel-Hashomer, Ramat-Gan 52621, Israel.,Sackler Faculty of Medicine, Tel Aviv University, Ramat Aviv, Tel Aviv 69978, Israel
| |
Collapse
|
28
|
Schierke F, Wyrwoll MJ, Wisdorf M, Niedzielski L, Maase M, Ruck T, Meuth SG, Kusche-Vihrog K. Nanomechanics of the endothelial glycocalyx contribute to Na +-induced vascular inflammation. Sci Rep 2017; 7:46476. [PMID: 28406245 PMCID: PMC5390251 DOI: 10.1038/srep46476] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2016] [Accepted: 03/20/2017] [Indexed: 12/11/2022] Open
Abstract
High dietary salt (NaCl) is a known risk factor for cardiovascular pathologies and inflammation. High plasma Na+ concentrations (high Na+) have been shown to stiffen the endothelial cortex and decrease nitric oxide (NO) release, a hallmark of endothelial dysfunction. Here we report that chronic high Na+ damages the endothelial glycocalyx (eGC), induces release of inflammatory cytokines from the endothelium and promotes monocyte adhesion. Single cell force spectroscopy reveals that high Na+ enhances vascular adhesion protein-1 (VCAM-1)-dependent adhesion forces between monocytes and endothelial surface, giving rise to increased numbers of adherent monocytes on the endothelial surface. Mineralocorticoid receptor antagonism with spironolactone prevents high Na+-induced eGC deterioration, decreases monocyte-endothelium interactions, and restores endothelial function, indicated by increased release of NO. Whereas high Na+ decreases NO release, it induces endothelial release of the pro-inflammatory cytokines IL-1ß and TNFα. However, in contrast to chronic salt load (hours), in vivo and in vitro, an acute salt challenge (minutes) does not impair eGC function. This study identifies the eGC as important mediator of inflammatory processes and might further explain how dietary salt contributes to endothelialitis and cardiovascular pathologies by linking endothelial nanomechanics with vascular inflammation.
Collapse
Affiliation(s)
- Florian Schierke
- Institute of Physiology II, University of Münster, 48149 Münster, Germany
| | - Margot J Wyrwoll
- Institute of Physiology II, University of Münster, 48149 Münster, Germany
| | - Martin Wisdorf
- Institute of Physiology II, University of Münster, 48149 Münster, Germany
| | - Leon Niedzielski
- Institute of Physiology II, University of Münster, 48149 Münster, Germany
| | - Martina Maase
- Institute of Physiology II, University of Münster, 48149 Münster, Germany
| | - Tobias Ruck
- Department of Neurology, University of Münster, 48149 Münster, Germany
| | - Sven G Meuth
- Department of Neurology, University of Münster, 48149 Münster, Germany
| | | |
Collapse
|
29
|
Binger KJ, Côrte-Real BF, Kleinewietfeld M. Immunometabolic Regulation of Interleukin-17-Producing T Helper Cells: Uncoupling New Targets for Autoimmunity. Front Immunol 2017; 8:311. [PMID: 28377767 PMCID: PMC5359241 DOI: 10.3389/fimmu.2017.00311] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2017] [Accepted: 03/06/2017] [Indexed: 01/04/2023] Open
Abstract
Interleukin-17-producing T helper (Th17) cells are critical for the host defense of bacterial and fungal pathogens and also play a major role in driving pathogenic autoimmune responses. Recent studies have indicated that the generation of Th17 cells from naïve CD4+ T cells is coupled with massive cellular metabolic adaptations, necessary to cope with different energy and metabolite requirements associated with switching from a resting to proliferative state. Furthermore, Th17 cells have to secure these metabolic adaptations when facing nutrient-limiting environments, such as at the sites of inflammation. Accumulating data indicates that this metabolic reprogramming is significantly linked to the differentiation of T helper cells and, particularly, that the metabolic changes of Th17 cells and anti-inflammatory Forkhead box P3+ regulatory T cells are tightly and reciprocally regulated. Thus, a better understanding of these processes could offer potential new targets for therapeutic interventions for autoimmune diseases. In this mini-review, we will highlight some of the recent advances and discoveries in the field, with a particular focus on metabolic demands of Th17 cells and their implications for autoimmunity.
Collapse
Affiliation(s)
- Katrina J Binger
- Department of Biochemistry and Molecular Biology, Bio21 Institute of Molecular Science and Biotechnology, University of Melbourne , Parkville, VIC , Australia
| | - Beatriz F Côrte-Real
- VIB Laboratory of Translational Immunomodulation, Hasselt University, BIOMED , Diepenbeek , Belgium
| | - Markus Kleinewietfeld
- VIB Laboratory of Translational Immunomodulation, Hasselt University, BIOMED , Diepenbeek , Belgium
| |
Collapse
|
30
|
Abstract
PURPOSE OF REVIEW Immune mechanisms exacerbate the severity of hypertension in humans and animal models of disease. This review summarizes recent mechanistic studies exploring the pathways whereby immunity influences salt-sensitive hypertension and renal disease. RECENT FINDINGS Emphasis is placed on the role of T cell subtypes, the mechanisms of T-cell activation, and the identification of potential antigens or neoantigens. SUMMARY Significant advancements have occurred in the search for pathways which activate the adaptive immune response. An enhanced understanding of the factors contributing to hypertension can lead to better therapies.
Collapse
|
31
|
Lou Y, Zhang F, Luo Y, Wang L, Huang S, Jin F. Serum and Glucocorticoid Regulated Kinase 1 in Sodium Homeostasis. Int J Mol Sci 2016; 17:ijms17081307. [PMID: 27517916 PMCID: PMC5000704 DOI: 10.3390/ijms17081307] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2016] [Revised: 08/02/2016] [Accepted: 08/03/2016] [Indexed: 12/13/2022] Open
Abstract
The ubiquitously expressed serum and glucocorticoid regulated kinase 1 (SGK1) is tightly regulated by osmotic and hormonal signals, including glucocorticoids and mineralocorticoids. Recently, SGK1 has been implicated as a signal hub for the regulation of sodium transport. SGK1 modulates the activities of multiple ion channels and carriers, such as epithelial sodium channel (ENaC), voltage-gated sodium channel (Nav1.5), sodium hydrogen exchangers 1 and 3 (NHE1 and NHE3), sodium-chloride symporter (NCC), and sodium-potassium-chloride cotransporter 2 (NKCC2); as well as the sodium-potassium adenosine triphosphatase (Na+/K+-ATPase) and type A natriuretic peptide receptor (NPR-A). Accordingly, SGK1 is implicated in the physiology and pathophysiology of Na+ homeostasis. Here, we focus particularly on recent findings of SGK1’s involvement in Na+ transport in renal sodium reabsorption, hormone-stimulated salt appetite and fluid balance and discuss the abnormal SGK1-mediated Na+ reabsorption in hypertension, heart disease, edema with diabetes, and embryo implantation failure.
Collapse
Affiliation(s)
- Yiyun Lou
- Department of Reproductive Endocrinology, Women's Hospital, School of Medicine, Zhejiang University, Hangzhou 310006, Zhejiang, China.
- Department of Gynaecology, Hangzhou Hospital of Traditional Chinese Medicine, Hangzhou 310007, Zhejiang, China.
| | - Fan Zhang
- Department of Reproductive Endocrinology, Women's Hospital, School of Medicine, Zhejiang University, Hangzhou 310006, Zhejiang, China.
| | - Yuqin Luo
- Department of Reproductive Endocrinology, Women's Hospital, School of Medicine, Zhejiang University, Hangzhou 310006, Zhejiang, China.
| | - Liya Wang
- Department of Reproductive Endocrinology, Women's Hospital, School of Medicine, Zhejiang University, Hangzhou 310006, Zhejiang, China.
| | - Shisi Huang
- Department of Reproductive Endocrinology, Women's Hospital, School of Medicine, Zhejiang University, Hangzhou 310006, Zhejiang, China.
| | - Fan Jin
- Department of Reproductive Endocrinology, Women's Hospital, School of Medicine, Zhejiang University, Hangzhou 310006, Zhejiang, China.
- Key Laboratory of Reproductive Genetics, National Ministry of Education (Zhejiang University), Women's Reproductive Healthy Laboratory of Zhejiang Province, Hangzhou 310058, Zhejiang, China.
| |
Collapse
|
32
|
Luo T, Ji WJ, Yuan F, Guo ZZ, Li YX, Dong Y, Ma YQ, Zhou X, Li YM. Th17/Treg Imbalance Induced by Dietary Salt Variation Indicates Inflammation of Target Organs in Humans. Sci Rep 2016; 6:26767. [PMID: 27353721 PMCID: PMC4926124 DOI: 10.1038/srep26767] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2016] [Accepted: 05/09/2016] [Indexed: 01/18/2023] Open
Abstract
The functions of T helper 17 (Th17) and regulatory T (Treg) cells are tightly orchestrated through independent differentiation pathways that are involved in the secretion of pro- and anti-inflammatory cytokines induced by high-salt dietary. However, the role of imbalanced Th17/Treg ratio implicated in inflammation and target organ damage remains elusive. Here, by flow cytometry analysis, we demonstrated that switching to a high-salt diet resulted in decreased Th17 cells and reciprocally increased Treg cells, leading to a decreased Th17/Treg ratio. Meanwhile, Th17-related pathway was down-regulated after one day of high salt loading, with the increase in high salt loading as shown by microarray and RT-PCR. Subsequently, blood oxygen level-dependent magnetic resonance imaging (BOLD-MRI) observed hypoxia in the renal medulla (increased R2* signal) during high-salt loading, which was regressed to its baseline level in a step-down fashion during low-salt feeding. The flow-mediated vasodilatation (FMD) of the branchial artery was significantly higher on the first day of high salt loading. Collectively, these observations indicate that a short-term increase in dietary salt intake could induce reciprocal switches in Th17/Treg ratio and related cytokines, which might be the underlying cellular mechanism of high-salt dietary induced end organ inflammation and potential atherosclerotic risk.
Collapse
Affiliation(s)
- Tao Luo
- Tianjin Key Laboratory of Cardiovascular Remodeling and Target Organ Injury, Pingjin Hospital Heart Center, Tianjin, P.R. China.,Department of Cardiology, No. 254 Hospital of PLA, Tianjin, P.R. China
| | - Wen-Jie Ji
- Tianjin Key Laboratory of Cardiovascular Remodeling and Target Organ Injury, Pingjin Hospital Heart Center, Tianjin, P.R. China
| | - Fei Yuan
- MRI Department, Pingjin Hospital, Tianjin, P.R. China
| | - Zhao-Zeng Guo
- Tianjin Key Laboratory of Cardiovascular Remodeling and Target Organ Injury, Pingjin Hospital Heart Center, Tianjin, P.R. China
| | - Yun-Xiao Li
- Tianjin Key Laboratory of Cardiovascular Remodeling and Target Organ Injury, Pingjin Hospital Heart Center, Tianjin, P.R. China
| | - Yan Dong
- Tianjin Key Laboratory of Cardiovascular Remodeling and Target Organ Injury, Pingjin Hospital Heart Center, Tianjin, P.R. China
| | - Yong-Qiang Ma
- Tianjin Key Laboratory of Cardiovascular Remodeling and Target Organ Injury, Pingjin Hospital Heart Center, Tianjin, P.R. China
| | - Xin Zhou
- Tianjin Key Laboratory of Cardiovascular Remodeling and Target Organ Injury, Pingjin Hospital Heart Center, Tianjin, P.R. China
| | - Yu-Ming Li
- Tianjin Key Laboratory of Cardiovascular Remodeling and Target Organ Injury, Pingjin Hospital Heart Center, Tianjin, P.R. China
| |
Collapse
|
33
|
Hypertension and immunity: mechanisms of T cell activation and pathways of hypertension. Curr Opin Nephrol Hypertens 2016; 24:470-4. [PMID: 26125645 DOI: 10.1097/mnh.0000000000000146] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
PURPOSE OF REVIEW The role of immune mechanisms to amplify hypertension in patients and animal models has been appreciated for decades. This review briefly summarizes recent studies exploring the mechanistic pathways, whereby the immune system participates in hypertension and renal disease. RECENT FINDINGS Emphasis in this review is placed upon recent studies exploring the role of T cell subtypes, newly described mechanisms of T cell activation, the identification of potential neoantigens, and environmental influences on immune cell activation. SUMMARY Significant advancements have been made in the search for antigens and pathways responsible for activation of the adaptive immune response, furthering our understanding of the factors contributing to hypertension and potentially leading to the development of new and more effective therapies.
Collapse
|
34
|
Abdoli A. Salt and miscarriage: Is there a link? Med Hypotheses 2016; 89:58-62. [PMID: 26968910 DOI: 10.1016/j.mehy.2016.02.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2015] [Revised: 01/21/2016] [Accepted: 02/03/2016] [Indexed: 11/27/2022]
Abstract
Salt is a major mineral element that plays fundamental roles in health and disease. Excessive salt intake is a major cause of hypertension, cardiovascular disease and stroke. Miscarriage and preeclampsia are the most common pregnancy complications with multiple etiological factors, including inflammatory and autoimmune conditions. More recently, different studies indicated that excessive salt intake is involved in the development of inflammatory processes through induction of T helper-17 pathway and their inflammatory cytokines. On the other hand, several studies indicated the pivotal role of inflammation in the etiology of miscarriage, preeclampsia and adverse pregnancy outcome. Here, it is hypothesized that excessive salt intake around the time of conception or during pregnancy can trigger inflammatory processes, which consequently associated with increased risk of miscarriage, preeclampsia or adverse pregnancy outcome. Thus, this hypothesis suggests that low salt intake around the time of conception or during pregnancy can decrease the risk of miscarriage or adverse pregnancy outcome. This hypothesis also offers new insights about the role of salt in the etiology of miscarriage and preeclampsia.
Collapse
Affiliation(s)
- Amir Abdoli
- Department of Parasitology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran.
| |
Collapse
|
35
|
Randell A, Daneshtalab N. Adjuvant-induced mono-arthritis potentiates cerebral hemorrhage in the spontaneously hypertensive rats. Life Sci 2016; 151:15-22. [PMID: 26903291 DOI: 10.1016/j.lfs.2016.02.012] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2015] [Revised: 01/14/2016] [Accepted: 02/04/2016] [Indexed: 11/28/2022]
Abstract
AIMS Patients with rheumatoid arthritis (RA), have a higher incidence of hypertension and stroke than the normal population. Currently there exists no animal model to study the pathogenic interactions of hemorrhagic stroke (HS) subsequent to chronic inflammation and hypertension. We have created and defined a hypertensive-mono-arthritic animal model who demonstrate gros signs of cerebral hemorrhage in presence of mono-arthritis. MAIN METHODS Spontaneously hypertensive rats (SHR) were fed either a high salt diet (4% NaCl; HSD) or Purina chow (RD) from weaning. Complete Freund's adjuvant (CFA) was injected into the left hind paw at 21-28weeks (control groups received saline (SAL)). Degree of inflammation, joint swelling, weight and blood pressure were monitored for 21days. Animals were then sacrificed and their brain and left hind paw evaluated. KEY FINDINGS All groups were hypertensive throughout the experimental period (>180mmHg systolic), irrespective of diet. Both CFA groups produced significant local inflammatory response in their injected paw with associated joint degradation and cellular infiltrates. Systemic plasma TNF-α levels were significantly elevated in CFA groups, with significant increase in TNF-α at 7 and 14days, compared to SAL groups. Cerebral hemorrhage was visualized in the CFA groups but not SAL controls, with a higher severity in HSD-CFA group. SIGNIFICANCE The mono-arthritic hypertensive animals are capable of developing HS upon induction of inflammatory insult. The HSD appears to exacerbate the inflammatory response and influence degree of the hemorrhage. Our novel, multi-disease model may provide an appropriate platform to study the pathogenesis of HS among arthritic patients.
Collapse
Affiliation(s)
- Amy Randell
- 300 Prince Philip Drive, Health Sciences Center, Memorial University of Newfoundland, School of Pharmacy, St. John's, Newfoundland A1B 3V6, Canada
| | - Noriko Daneshtalab
- 300 Prince Philip Drive, Health Sciences Center, Memorial University of Newfoundland, School of Pharmacy, St. John's, Newfoundland A1B 3V6, Canada.
| |
Collapse
|
36
|
Hucke S, Eschborn M, Liebmann M, Herold M, Freise N, Engbers A, Ehling P, Meuth SG, Roth J, Kuhlmann T, Wiendl H, Klotz L. Sodium chloride promotes pro-inflammatory macrophage polarization thereby aggravating CNS autoimmunity. J Autoimmun 2015; 67:90-101. [PMID: 26584738 DOI: 10.1016/j.jaut.2015.11.001] [Citation(s) in RCA: 117] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2015] [Revised: 10/23/2015] [Accepted: 11/02/2015] [Indexed: 12/13/2022]
Abstract
The increasing incidence in Multiple Sclerosis (MS) during the last decades in industrialized countries might be linked to a change in dietary habits. Nowadays, enhanced salt content is an important characteristic of Western diet and increased dietary salt (NaCl) intake promotes pathogenic T cell responses contributing to central nervous system (CNS) autoimmunity. Given the importance of macrophage responses for CNS disease propagation, we addressed the influence of salt consumption on macrophage responses in CNS autoimmunity. We observed that EAE-diseased mice receiving a NaCl-high diet showed strongly enhanced macrophage infiltration and activation within the CNS accompanied by disease aggravation during the effector phase of EAE. NaCl treatment of macrophages elicited a strong pro-inflammatory phenotype characterized by enhanced pro-inflammatory cytokine production, increased expression of immune-stimulatory molecules, and an antigen-independent boost of T cell proliferation. This NaCl-induced pro-inflammatory macrophage phenotype was accompanied by increased activation of NF-kB and MAPK signaling pathways. The pathogenic relevance of NaCl-conditioned macrophages is illustrated by the finding that transfer into EAE-diseased animals resulted in significant disease aggravation compared to untreated macrophages. Importantly, also in human monocytes, NaCl promoted a pro-inflammatory phenotype that enhanced human T cell proliferation. Taken together, high dietary salt intake promotes pro-inflammatory macrophages that aggravate CNS autoimmunity. Together with other studies, these results underline the need to further determine the relevance of increased dietary salt intake for MS disease severity.
Collapse
Affiliation(s)
| | | | - Marie Liebmann
- Department of Neurology, University of Muenster, Germany
| | - Martin Herold
- Department of Neurology, University of Muenster, Germany
| | - Nicole Freise
- Institute of Immunology, University of Muenster, Germany
| | - Annika Engbers
- Department of Neurology, University of Muenster, Germany
| | - Petra Ehling
- Department of Neurology, University of Muenster, Germany
| | - Sven G Meuth
- Department of Neurology, University of Muenster, Germany; Cells in Motion, Cluster of Excellence, University of Münster, Germany
| | - Johannes Roth
- Institute of Immunology, University of Muenster, Germany
| | - Tanja Kuhlmann
- Institute of Neuropathology, University of Muenster, Germany
| | - Heinz Wiendl
- Department of Neurology, University of Muenster, Germany; Cells in Motion, Cluster of Excellence, University of Münster, Germany
| | - Luisa Klotz
- Department of Neurology, University of Muenster, Germany.
| |
Collapse
|
37
|
Hernandez AL, Kitz A, Wu C, Lowther DE, Rodriguez DM, Vudattu N, Deng S, Herold KC, Kuchroo VK, Kleinewietfeld M, Hafler DA. Sodium chloride inhibits the suppressive function of FOXP3+ regulatory T cells. J Clin Invest 2015; 125:4212-22. [PMID: 26524592 DOI: 10.1172/jci81151] [Citation(s) in RCA: 257] [Impact Index Per Article: 25.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2015] [Accepted: 08/20/2015] [Indexed: 12/12/2022] Open
Abstract
FOXP3+ Tregs are central for the maintenance of self-tolerance and can be defective in autoimmunity. In multiple sclerosis and type-1 diabetes, dysfunctional self-tolerance is partially mediated by a population of IFNγ-secreting Tregs. It was previously reported that increased NaCl concentrations promote the induction of proinflammatory Th17 cells and that high-salt diets exacerbate experimental models of autoimmunity. Here, we have shown that increasing NaCl, either in vitro or in murine models via diet, markedly impairs Treg function. NaCl increased IFNγ secretion in Tregs, and reducing IFNγ - either by neutralization with anti-IFNγ antibodies or shRNA-mediated knockdown - restored suppressive activity in Tregs. The heightened IFNγ secretion and loss of Treg function were mediated by the serum/glucocorticoid-regulated kinase (SGK1). A high-salt diet also impaired human Treg function and was associated with the induction of IFNγ-secreting Tregs in a xenogeneic graft-versus-host disease model and in adoptive transfer models of experimental colitis. Our results demonstrate a putative role for an environmental factor that promotes autoimmunity by inducing proinflammatory responses in CD4 effector cells and Treg pathways.
Collapse
MESH Headings
- Adoptive Transfer
- Animals
- Antibodies, Neutralizing/immunology
- Autoimmunity/drug effects
- CD4-Positive T-Lymphocytes/immunology
- Cells, Cultured
- Coculture Techniques
- Colitis/immunology
- Cytokines/biosynthesis
- Cytokines/genetics
- Forkhead Transcription Factors/analysis
- Forkhead Transcription Factors/genetics
- Gene Expression Profiling
- Genes, Reporter
- Graft vs Host Disease/immunology
- Heterografts
- Humans
- Immediate-Early Proteins/physiology
- Inflammation
- Interferon-gamma/genetics
- Interferon-gamma/metabolism
- Interferon-gamma/pharmacology
- Leukocytes, Mononuclear/transplantation
- Male
- Mice
- Protein Serine-Threonine Kinases/physiology
- RNA Interference
- RNA, Small Interfering/genetics
- Sodium Chloride/pharmacology
- Sodium Chloride, Dietary/adverse effects
- Sodium Chloride, Dietary/pharmacology
- T-Lymphocytes, Regulatory/drug effects
- T-Lymphocytes, Regulatory/immunology
- T-Lymphocytes, Regulatory/metabolism
Collapse
|
38
|
Hofmeister LH, Perisic S, Titze J. Tissue sodium storage: evidence for kidney-like extrarenal countercurrent systems? Pflugers Arch 2015; 467:551-8. [PMID: 25600900 DOI: 10.1007/s00424-014-1685-x] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2014] [Accepted: 12/29/2014] [Indexed: 11/29/2022]
Abstract
Recent evidence from chemical analysis of tissue electrolyte and water composition has shown that body Na(+) content in experimental animals is not constant, does not always readily equilibrate with water, and cannot be exclusively controlled by the renal blood purification process. Instead, large amounts of Na(+) are stored in the skin and in skeletal muscle. Quantitative non-invasive detection of Na(+) reservoirs with sodium magnetic resonance imaging ((23)NaMRI) suggests that this mysterious Na(+) storage is not only an animal research curiosity but also exists in humans. In clinical studies, tissue Na(+) storage is closely associated with essential hypertension. In animal experiments, modulation of reservoir tissue Na(+) content leads to predictable blood pressure changes. The available evidence thus suggests that the patho(?)-physiological process of Na(+) storage might be of relevance for human health and disease.
Collapse
Affiliation(s)
- Lucas H Hofmeister
- Division of Clinical Pharmacology, Vanderbilt University School of Medicine, 2213 Garland Avenue, P435F Medical Research Building IV, Nashville, TN, 37232, USA
| | | | | |
Collapse
|