1
|
Al-Mhanna SB, Franklin BA, Jakicic JM, Stamatakis E, Pescatello LS, Riebe D, Thompson WR, Skinner J, Colberg SR, Alkhamees NH, Bin Sheeha B, Gülü M, Alghannam AF, Batrakoulis A. Impact of resistance training on cardiometabolic health-related indices in patients with type 2 diabetes and overweight/obesity: a systematic review and meta-analysis of randomised controlled trials. Br J Sports Med 2025; 59:733-746. [PMID: 39773835 DOI: 10.1136/bjsports-2024-108947] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/12/2024] [Indexed: 01/11/2025]
Abstract
OBJECTIVE To evaluate the effects of resistance training on cardiometabolic health-related outcomes in patients with type 2 diabetes mellitus (T2DM) and overweight/obesity. DESIGN Systematic review and meta-analysis of randomised controlled trials (RCTs). DATA SOURCES PubMed, Web of Science, Scopus, Science Direct, Cochrane Library and Google Scholar databases were searched from inception up to May 2024. The search strategy included the following keywords: diabetes, resistance exercise and strength training. ELIGIBILITY CRITERIA FOR SELECTING STUDIES RCTs published in English comparing resistance training alone with non-exercising standard treatment. Participants were adults diagnosed with T2DM and concurrent overweight/obesity (body mass index (BMI) ≥25 kg/m2). RESULTS A total of 18 RCTs qualified involving 1180 patients (48.6/51.4 female/male ratio; 63.3±7.0 years; 29.3±4.3 kg/m2). Waist circumference (standardised mean differences (SMD) -0.85 cm, 95% CI -1.66 to -0.04), waist-to-hip ratio (SMD -0.72, 95% CI -1.30 to -0.15), high-density lipoprotein cholesterol (SMD +0.40 mg/dL, 95% CI 0.07 to -0.72), triglycerides (SMD -0.54 mg/dL, 95% CI -1.06 to -0.02), fasting blood glucose (SMD -0.65 mmol/L, 95% CI -1.19 to -0.12), fasting insulin (SMD -0.74 uIU/mL, 95% CI -1.12 to -0.36) and glycated haemoglobin (SMD -0.32%, 95% CI -0.63 to -0.01) improved compared with standard treatment. The risk of bias was low to unclear, and the quality of evidence was very low to moderate. CONCLUSIONS Resistance training as a standalone exercise intervention in the management and treatment of T2DM with concurrent overweight/obesity is associated with many cardiometabolic benefits when compared with standard treatment without exercise. PROSPERO REGISTRATION NUMBER CRD42022355612.
Collapse
Affiliation(s)
- Sameer Badri Al-Mhanna
- Center for Global Health Research, Saveetha Institute of Medical and Technical Sciences (Deemed to be University), Chennai, Tamil Nadu, India
- Department of Higher Studies, Al Qasim Green University, Al Qasim, Iraq
| | - Barry A Franklin
- Preventive Cardiology and Cardiac Rehabilitation, Corewell Health Beaumont Hospital, Royal Oak, Michigan, USA
- Internal Medicine, Oakland University William Beaumont School of Medicine, Rochester, Michigan, USA
| | - John M Jakicic
- Department of Internal Medicine, The University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Emmanuel Stamatakis
- Mackenzie Wearables Research Hub, Charles Perkins Centre, The University of Sydney, Syndey, New South Wales, Australia
- Faculty of Medicine and Health, School of Health Sciences, The University of Sydney, Sydney, New South Wales, Australia
| | - Linda S Pescatello
- Department of Kinesiology, University of Connecticut, Storrs, Connecticut, USA
| | - Deborah Riebe
- College of Health Sciences, University of Rhode Island, Kingston, Rhode Island, USA
| | - Walter R Thompson
- 8College of Education and Human Development, Georgia State University, Atlanta, Georgia, USA
| | - James Skinner
- Department of Kinesiology, Indiana University Bloomington, Bloomington, Indiana, USA
| | - Sheri R Colberg
- Human Movement Sciences Department, Old Dominion University, Norfolk, Virginia, USA
| | - Nouf H Alkhamees
- Department of Rehabilitation, Princess Nourah bint Abdulrahman University, Riyadh, Saudi Arabia
| | - Bodor Bin Sheeha
- Department of Rehabilitation, Princess Nourah bint Abdulrahman University, Riyadh, Saudi Arabia
| | - Mehmet Gülü
- Department of Sports Management, Kirikkale University, Kirikkale, Turkey
| | - Abdullah F Alghannam
- Lifestyle and Health Research Center, Princess Nourah bint Abdulrahman University, Riyadh, Saudi Arabia
| | - Alexios Batrakoulis
- Department of Physical Education and Sport Science, University of Thessaly, Trikala, Greece
- Department of Physical Education and Sport Science, Democritus University of Thrace, Komotini, Greece
| |
Collapse
|
2
|
Zheng XY, Chen Y, Gao Y, Zhou M, Guan WZ. Association between Dietary Inflammatory Index (DII) and sarcopenia in ischemic heart disease: an 1999-2004 NHANES study of 1088 US individuals. Minerva Cardiol Angiol 2025; 73:65-72. [PMID: 38804626 DOI: 10.23736/s2724-5683.24.06496-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/29/2024]
Abstract
BACKGROUND The aim of this study was to investigate the relationship between DII and sarcopenia in individuals with ischemic heart disease (IHD). METHODS This was a retrospective study utilizing data of the National Health and Nutrition Examination Survey (NHANES) from 1999-2004. Adults aged ≥50 years diagnosed with IHD, having complete 24-hour dietary recall data, and dual energy X-ray absorptiometry (DEXA)-measured muscle mass were eligible for inclusion. Association between DII and sarcopenia, defined by reduced appendicular skeletal muscle mass, was determined by the logistic regression analyses. RESULTS Data of 1088 individuals were analyzed, with the mean age of 68.1±0.5 years. Significantly higher DII was observed in the sarcopenic group compared to the non-sarcopenic group (0.24 vs. -0.17, P=0.020). After adjusting for relevant confounders in the multivariable analysis, each unit increase in DII was significantly associated with higher odds of sarcopenia (adjusted odd ratio [aOR]=1.07, 95% confidence interval: 1.00-1.14, P value = 0.040). In stratified analyses, among patients with a Body Mass Index (BMI) ≥30 kg/m2, both DII tertile 2 and tertile 3 were significantly associated with greater odds of sarcopenia (tertile 2 vs. tertile 1: aOR=2.85, 95% CI: 1.56-5.23, P=0.001; tertile 3 vs. tertile 1: aOR=3.11, 95% CI: 1.53-6.31, P=0.002), whereas no significant associations was observed among patients with a BMI<30 kg/m2. CONCLUSIONS This study has established a significant independent association between a higher DII and an increased risk of sarcopenia in US adults with IHD regardless of type of IHD. BMI appears as a moderating factor in this association.
Collapse
Affiliation(s)
- Xiao-Yun Zheng
- Department of Health Care, China-Japan Friendship Hospital, Beijing, China -
| | - Yi Chen
- Department of Health Care, China-Japan Friendship Hospital, Beijing, China
| | - Yan Gao
- Department of Health Care, China-Japan Friendship Hospital, Beijing, China
| | - Mo Zhou
- Department of Health Care, China-Japan Friendship Hospital, Beijing, China
| | - Wen-Zhuo Guan
- Department of Health Care, China-Japan Friendship Hospital, Beijing, China
| |
Collapse
|
3
|
Li J, Zhao S, Zhang X, Fan M, Wan J, Lin R, Fan F, Liu G, Guan S, Liu A. Association of Systemic Inflammatory Response Index with the cardiometabolic multimorbidity among US adults: A population-based study. Brain Circ 2025; 11:39-47. [PMID: 40224555 PMCID: PMC11984816 DOI: 10.4103/bc.bc_32_24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Revised: 11/08/2024] [Accepted: 11/22/2024] [Indexed: 04/15/2025] Open
Abstract
BACKGROUND Chronic inflammation plays an essential role in the occurrence and progression of cardiometabolic diseases (CMDs). We aim to examine the association between a novel inflammatory biomarker Systemic Inflammatory Response Index (SIRI) and different cardiometabolic multimorbidity (CMM) statuses. METHODS This was a cross-sectional study that includes general participants of the National Health and Nutrition Examination Survey database from 1999 to 2018. SIRI was calculated as neutrophil count × lymphocyte count/monocyte count. The CMDs were defined as a series of diseases including diabetes mellitus (DM), heart disease (HD), and stroke. We explored the association of SIRI with outcomes with weighted multivariable logistic regression models weighted restricted cubic spline. The diagnostic value of SIRI was evaluated using weighted receiver operating characteristic (ROC) curves. RESULTS A total of 43,345 participants were enrolled with a mean age of 45.86 years. The weighted prevalence of CMD and CMM was 17.14% and 2.94%, respectively. Compared to those without CMD, the adjusted odds ratios (95% confidence interval) for each unit increase in SIRI were 1.14 (1.09-1.19) for DM, 1.13 (1.07-1.19) for HD, 1.11 (1.04-1.19) for stroke, 1.17 (1.12-1.22) for CMD, and 1.16 (1.10-1.23) for CMM, according to the weighted multivariable logistic regression. Elevated SIRI level was independently associated with increased CMM. There was no interaction found in subgroup analysis. According to the ROC analysis, SIRI had a superior diagnostic ability to neutrophil-lymphocyte ratio, platelet-lymphocyte ratio, and monocyte-lymphocyte ratio for CMD (area under the curve [AUC] =0.581) and CMM (AUC = 0.633). CONCLUSIONS Elevated level of SIRI was positively associated with the prevalence of DM, coronary artery disease, stroke, CMD, and CMM, suggesting that SIRI could be a potential noninvasive biomarker for CMD and CMM.
Collapse
Affiliation(s)
- Jing Li
- Department of Neurointervention at The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Songfeng Zhao
- Department of Neurosurgery, the Third Xiangya Hospital, Central South University, Beijing, China
| | - Xiaowen Zhang
- Department of Neurointervention at The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Mengyan Fan
- Department of Neurointervention at The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Jiaxin Wan
- Department of Neurointervention at The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Rijin Lin
- Department of Neurointervention at The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Feng Fan
- Department of Neurointervention at The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Guo Liu
- Department of Neurointervention at The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Sheng Guan
- Department of Neurointervention at The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Aihua Liu
- Department of Neurointervention, Beijing Neurosurgical Institute, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
4
|
Han L, Gan Y, Du J, Hu Y, Chen Y, Huang Q, Zhang Z, Yawalkar N, Yan K, Wang Z. Evaluation of β2-microglobulin in the condition and prognosis of psoriasis patients. J DERMATOL TREAT 2024; 35:2377665. [PMID: 39069294 DOI: 10.1080/09546634.2024.2377665] [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: 04/24/2024] [Accepted: 07/03/2024] [Indexed: 07/30/2024]
Abstract
BACKGROUND Numerous studies have linked the inflammatory pathway in psoriasis and metabolic disease, while no specific marker defined it. It is worth exploring the association of β2-microglobulin (β2M) in psoriasis severity and comorbidities. OBJECTIVES To investigate the correlation between blood β2M level and psoriasis severity, to explore the inflammatory factors influencing the occurrence of psoriasis comorbidities such as arthritis, diabetes, and hypertension. METHODS Ninety-seven psoriasis patients were analyzed in the cohort retrospective study during 12 weeks. RESULTS Significantly higher levels of blood β2M and ESR were observed in the group that patients' PASI ≥10 than in the group that PASI <10. Blood β2M level had strong significantly positive correlations with the PASI in Pearson's correlation analysis. In the model that systemic inflammatory factors to find psoriasis comorbidity risk factors, logistic regression analysis showed that blood β2M level was the significant risk factor associated with diabetes and hypertension. High-sensitivity C-reactive protein (hsCRP) was the significant risk factor associated with arthritis. CONCLUSIONS Patients with a severer psoriasis tended to have higher blood β2M levels and severer inflammatory state. In the systemic inflammation indexes, the level of blood β2M affected the risk of hypertension and diabetes, and hsCRP affected the risk of arthritis in patients with psoriasis.
Collapse
Affiliation(s)
- Ling Han
- Department of Dermatology, Huashan Hospital, Fudan University, Shanghai Institute of Dermatology, Shanghai, PR China
| | - Yixiao Gan
- Department of Transfusion Medicine, Huashan Hospital, Fudan University, Shanghai, China
| | - Juan Du
- Department of Dermatology, Huashan Hospital, Fudan University, Shanghai Institute of Dermatology, Shanghai, PR China
| | - Yao Hu
- Department of Laboratory Medicine, Huashan Hospital, Fudan University, Shanghai, China
| | - Yanwen Chen
- Department of Laboratory Medicine, Huashan Hospital, Fudan University, Shanghai, China
| | - Qiong Huang
- Department of Dermatology, Huashan Hospital, Fudan University, Shanghai Institute of Dermatology, Shanghai, PR China
| | - Zhenghua Zhang
- Department of Dermatology, Huashan Hospital, Fudan University, Shanghai Institute of Dermatology, Shanghai, PR China
| | - Nikhil Yawalkar
- Department of Dermatology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Kexiang Yan
- Department of Dermatology, Huashan Hospital, Fudan University, Shanghai Institute of Dermatology, Shanghai, PR China
| | - Zhicheng Wang
- Department of Transfusion Medicine, Huashan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
5
|
Chen Z, Wu J, Ai K, Bu Z, Niu W, Li M. Trends in Children's Dietary Inflammatory Index and association with prediabetes in U.S. adolescents. Nutr Diabetes 2024; 14:94. [PMID: 39557855 PMCID: PMC11574316 DOI: 10.1038/s41387-024-00349-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/18/2024] [Revised: 10/23/2024] [Accepted: 10/30/2024] [Indexed: 11/20/2024] Open
Abstract
BACKGROUND AND OBJECTIVES Prediabetes is a high-risk state for diabetes. We aimed to illustrate secular trends in the Children's Dietary Inflammation Index (C-DII) among U.S. adolescents and assess its association with prediabetes. METHODS Adolescents aged 12-18 years were collected from the National Health and Nutrition Examination Survey, 2001-2018. Prediabetes was defined based on Hemoglobin A1c, fasting glucose, and glucose tolerance levels. Risk was quantified by odds ratio (OR) and 95% confidence interval (CI). RESULTS A total of 13,684 adolescents were analyzed, representing a weighted total population of 33,351,181. C-DII scores declined significantly from 2001 to 2012 and increased from 2013 to 2018. The relationship between C-DII and prediabetes was roughly linear. When assigning the low C-DII scores as the reference, adolescents with medium and high C-DII scores were 1.22 (adjusted 95% CI: 1.04-1.44) and 1.25 (0.99-1.60) times more likely to have prediabetes. In subgroup analyses, the risk for prediabetes was significantly enhanced in boys (adjusted OR = 1.26 and 1.45 for medium and high C-DII scores, 95% CI: 1.05-1.51 and 1.09-1.92), and in adolescents living in poor families for medium (1.34 and 1.44, 1.08-1.67 and 1.07-1.95). CONCLUSIONS Our findings indicate a V-shaped secular trend in C-DII scores from 2001 to 2018 in U.S. adolescents, with the nadir in 2011-2012, and the risk for prediabetes was significantly increased by over 20% in adolescents possessing medium or high C-DII scores.
Collapse
Affiliation(s)
- Zisu Chen
- Department of Pediatrics, Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, China
| | - Jing Wu
- Center for Evidence-Based Medicine, Capital Institute of Pediatrics, Beijing, China
| | - Kepeng Ai
- Department of Gastroenterology, Yan'an University Affiliated Hospital, Shaanxi, China
| | - Zhuying Bu
- Department of Gastroenterology, Yan'an University Affiliated Hospital, Shaanxi, China
| | - Wenquan Niu
- Center for Evidence-Based Medicine, Capital Institute of Pediatrics, Beijing, China
| | - Min Li
- Department of Pediatrics, Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, China.
| |
Collapse
|
6
|
Xu Q, Tian X, Xia X, Zhang Y, Zheng M, Wang A. Estimated glucose disposal rate, high sensitivity C-reactive protein and cardiometabolic multimorbidity in middle-aged and older Chinese adults: A nationwide prospective cohort study. Diabetes Res Clin Pract 2024; 217:111894. [PMID: 39414087 DOI: 10.1016/j.diabres.2024.111894] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/13/2024] [Revised: 10/09/2024] [Accepted: 10/13/2024] [Indexed: 10/18/2024]
Abstract
AIM To explore the separate and joint association of estimated glucose disposal rate (eGDR) and high-sensitivity C-reactive protein (hsCRP) with cardiometabolic multimorbidity (CMM). METHODS A total of 6900 participants aged 45 years or older with available data on eGDR and hsCRP and without cardiometabolic diseases at baseline from the China Health and Retirement Longitudinal Study were included. CMM was defined as the coexistence of two or more cardiometabolic diseases, including heart diseases, stroke, and diabetes. RESULTS During a median follow-up of 9.0 years, 464 (6.7 %) participants developed CMM. Low eGDR and high hsCRP separately and jointly increased the risk of CMM. The adjusted hazard ratio (HR) was 1.67 (95 % confidence interval [CI] 1.33-2.09) for low eGDR versus high eGDR, 1.43 (95 % CI 1.12-1.82) for high hsCRP versus low hsCRP) and 2.40 (95 % CI 1.77-3.27) for low eGDR plus high hsCRP versus high eGDR plus low hsCRP. The C-statistic, discriminatory power and risk reclassification significantly improved with the addition of combined eGDR and hsCRP for CMM (P < 0.001). CONCLUSIONS Low eGDR and high hsCRP were individually and jointly associated with increased risk of incident CMM. The findings highlighted the importance of joint evaluation of eGDR and hsCRP for primary prevention of CMM.
Collapse
Affiliation(s)
- Qin Xu
- Department of Epidemiology, Beijing Neurosurgical Institute, Beijing Tiantan Hospital, Capital Medical University, Beijing, China; China National Clinical Research Center for Neurological Diseases, Beijing Tiantan Hospital, Capital Medical University, Beijing, China; Department of Clinical Epidemiology and Clinical Trial, Capital Medical University, Beijing, China
| | - Xue Tian
- Department of Epidemiology and Health Statistics, School of Public Health, Capital Medical University, Beijing, China
| | - Xue Xia
- Department of Epidemiology, Beijing Neurosurgical Institute, Beijing Tiantan Hospital, Capital Medical University, Beijing, China; China National Clinical Research Center for Neurological Diseases, Beijing Tiantan Hospital, Capital Medical University, Beijing, China; Department of Clinical Epidemiology and Clinical Trial, Capital Medical University, Beijing, China
| | - Yijun Zhang
- Department of Epidemiology and Health Statistics, School of Public Health, Capital Medical University, Beijing, China
| | - Manqi Zheng
- Department of Epidemiology, Beijing Neurosurgical Institute, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Anxin Wang
- Department of Epidemiology, Beijing Neurosurgical Institute, Beijing Tiantan Hospital, Capital Medical University, Beijing, China; China National Clinical Research Center for Neurological Diseases, Beijing Tiantan Hospital, Capital Medical University, Beijing, China; Department of Clinical Epidemiology and Clinical Trial, Capital Medical University, Beijing, China.
| |
Collapse
|
7
|
Bernatova I, Bartekova M. Molecular Aspects of Cardiometabolic Diseases: From Etiopathogenesis to Potential Therapeutic Targets. Int J Mol Sci 2024; 25:5841. [PMID: 38892029 PMCID: PMC11172306 DOI: 10.3390/ijms25115841] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Accepted: 05/23/2024] [Indexed: 06/21/2024] Open
Abstract
Cardiometabolic diseases (CMDs) encompass a range of prevalent, often preventable, non-communicable illnesses, including myocardial infarction, stroke, cardiac insufficiency, arterial hypertension, obesity, type 2 diabetes mellitus, insulin resistance, chronic renal dysfunction, non-alcoholic fatty liver disease, and rare metabolic disorders [...].
Collapse
Affiliation(s)
- Iveta Bernatova
- Institute of Normal and Pathological Physiology, Centre of Experimental Medicine, Slovak Academy of Sciences, Sienkiewiczova 1, 81371 Bratislava, Slovakia
| | - Monika Bartekova
- Institute for Heart Research, Centre of Experimental Medicine, Slovak Academy of Sciences, 9 Dubravska cesta, 84104 Bratislava, Slovakia
| |
Collapse
|
8
|
Kamurai B, Chikwati RP, Vhanda D, Nyamayaro T, Manasa J, Kouamou V. Effect of dolutegravir on ferritin, iron, and C-reactive protein among people living with HIV and co-infections. South Afr J HIV Med 2024; 25:1543. [PMID: 38725704 PMCID: PMC11079419 DOI: 10.4102/sajhivmed.v25i1.1543] [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: 10/09/2023] [Accepted: 11/28/2023] [Indexed: 05/12/2024] Open
Abstract
Background Dolutegravir-based antiretroviral therapy (ART) is currently recommended as the preferred first-line ART in many resource-limited settings. However, little is known about the clinical experience of dolutegravir within a context of prevalent co-infections. Objectives To assess virological outcomes, and iron, ferritin and C-reactive protein (CRP) levels among people living with HIV (PLWH) and co-infections after initiating or re-initiating dolutegravir-based ART. Method This prospective study was conducted between August 2022 and August 2023. Study participants were recruited from an HIV opportunistic infection clinic. Screening for co-infections (syphilis, hepatitis B virus, cytomegalovirus and herpes simplex virus) was performed at baseline, prior to ART initiation. Plasma HIV viral load (VL), CRP, ferritin and iron levels were measured at baseline and at the 6-month follow-up period. Results A total of 100 participants (51 women and 49 men) were enrolled in this study. The median age of the participants was 39 years. The prevalence of co-infections was 30%. Prior to ART initiation, participants with co-infections had higher VL, CRP and ferritin, and lower iron levels, compared to those without co-infections (P < 0.001). Following 6 months of ART, CRP and ferritin levels decreased while iron levels increased, regardless of co-infection status. However, CRP and ferritin remained significantly higher in those with co-infections despite similar and high rates of virologic suppression in both groups. Conclusion The presence of co-infections in PLWH is associated with higher VL and with chronic inflammation. Ferritin and CRP decreased on dolutegravir-based ART but remained higher in people with co-infections despite similar rates of virologic suppression.
Collapse
Affiliation(s)
- Bridget Kamurai
- Department of Chemical Pathology, Faculty of Medicine and Health Sciences, University of Zimbabwe, Harare, Zimbabwe
| | - Raylton P. Chikwati
- Department of Chemical Pathology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
- Sydney Brenner Institute for Molecular Bioscience, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Donald Vhanda
- Department of Chemical Pathology, Faculty of Medicine and Health Sciences, University of Zimbabwe, Harare, Zimbabwe
| | - Terrence Nyamayaro
- Department of Chemical Pathology, Faculty of Medicine and Health Sciences, University of Zimbabwe, Harare, Zimbabwe
| | - Justen Manasa
- Department of Internal Medicine, Faculty of Medicine and Health Sciences, University of Zimbabwe, Harare, Zimbabwe
| | - Vinie Kouamou
- Department of Internal Medicine, Faculty of Medicine and Health Sciences, University of Zimbabwe, Harare, Zimbabwe
| |
Collapse
|
9
|
Deng W, Yan Y, Shi C, Sui D. Single-cell and bulk RNAseq unveils the immune infiltration landscape and targeted therapeutic biomarkers of psoriasis. Front Genet 2024; 15:1365273. [PMID: 38699235 PMCID: PMC11063342 DOI: 10.3389/fgene.2024.1365273] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Accepted: 04/01/2024] [Indexed: 05/05/2024] Open
Abstract
Background Psoriasis represents a multifaceted and debilitating immune-mediated systemic ailment afflicting millions globally. Despite the continuous discovery of biomarkers associated with psoriasis, identifying lysosomal biomarkers, pivotal as cellular metabolic hubs, remains elusive. Methods We employed a combination of differential expression analysis and weighted gene co-expression network analysis (WGCNA) to initially identify lysosomal genes. Subsequently, to mitigate overfitting and eliminate collinear genes, we applied 12 machine learning algorithms to screen robust lysosomal genes. These genes underwent further refinement through random forest (RF) and Lasso algorithms to ascertain the final hub lysosomal genes. To assess their predictive efficacy, we conducted receiver operating characteristic (ROC) analysis and verified the expression of diagnostic biomarkers at both bulk and single-cell levels. Furthermore, we utilized single-sample gene set enrichment analysis (ssGSEA), CIBERSORT, and Pearson's correlation analysis to elucidate the association between immune phenotypes and hub lysosomal genes in psoriatic samples. Finally, employing the Cellchat algorithm, we explored potential mechanisms underlying the participation of these hub lysosomal genes in cell-cell communication. Results Functional enrichment analyses revealed a close association between psoriasis and lysosomal functions. Subsequent intersection analysis identified 19 key lysosomal genes, derived from DEGs, phenotypic genes of WGCNA, and lysosomal gene sets. Following the exclusion of collinear genes, we identified 11 robust genes, further refined through RF and Lasso, yielding 3 hub lysosomal genes (S100A7, SERPINB13, and PLBD1) closely linked to disease occurrence, with high predictive capability for disease diagnosis. Concurrently, we validated their relative expression in separate bulk datasets and single-cell datasets. A nomogram based on these hub genes may offer clinical advantages for patients. Notably, these three hub genes facilitated patient classification into two subtypes, namely metabolic-immune subtype 1 and signaling subtype 2. CMap analysis suggested butein and arachidonic fasudil as preferred treatment agents for subtype 1 and subtype 2, respectively. Finally, through Cellchat and correlation analysis, we identified PRSS3-F2R as potentially promoting the expression of hub genes in the psoriasis group, thereby enhancing keratinocyte-fibroblast interaction, ultimately driving psoriasis occurrence and progression. Conclusion Our study identifies S100A7, SERPINB13, and PLBD1 as potential diagnostic biomarkers, offering promising prospects for more precisely tailored psoriatic immunotherapy designs.
Collapse
Affiliation(s)
- Wenhui Deng
- The First Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
- The First Affiliated Hospital of Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Yijiao Yan
- The First Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
- The First Affiliated Hospital of Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Chengzhi Shi
- The First Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
- The First Affiliated Hospital of Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Daoshun Sui
- The First Affiliated Hospital of Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| |
Collapse
|
10
|
Suffee N, Le Goff W, Chen J. Editorial: Cardiometabolic diseases and inflammatory responses. Front Immunol 2024; 15:1384022. [PMID: 38495875 PMCID: PMC10940500 DOI: 10.3389/fimmu.2024.1384022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2024] [Accepted: 02/13/2024] [Indexed: 03/19/2024] Open
Affiliation(s)
- Nadine Suffee
- Sorbonne Université, INSERM, Foundation for Innovation in Cardiometabolism and Nutrition (ICAN), UMR_S1166, Paris, France
| | - Wilfried Le Goff
- Sorbonne Université, INSERM, Foundation for Innovation in Cardiometabolism and Nutrition (ICAN), UMR_S1166, Paris, France
| | - Jianmin Chen
- William Harvey Research Institute, Queen Mary University of London, London, United Kingdom
- Centre for inflammation and Therapeutic Innovation, Queen Mary University of London, London, United Kingdom
| |
Collapse
|
11
|
Chiș A, Noubissi PA, Pop OL, Mureșan CI, Fokam Tagne MA, Kamgang R, Fodor A, Sitar-Tăut AV, Cozma A, Orășan OH, Hegheș SC, Vulturar R, Suharoschi R. Bioactive Compounds in Moringa oleifera: Mechanisms of Action, Focus on Their Anti-Inflammatory Properties. PLANTS (BASEL, SWITZERLAND) 2023; 13:20. [PMID: 38202328 PMCID: PMC10780634 DOI: 10.3390/plants13010020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Revised: 12/06/2023] [Accepted: 12/15/2023] [Indexed: 01/12/2024]
Abstract
Moringa oleifera (M. oleifera) is a tropical tree native to Pakistan, India, Bangladesh, and Afghanistan; it is cultivated for its nutritious leaves, pods, and seeds. This scientific study was conducted to outline the anti-inflammatory properties and mechanisms of action of bioactive compounds from M. oleifera. The existing research has found that the plant is used in traditional medicine due to its bioactive compounds, including phytochemicals: flavonoids and polyphenols. The compounds are thought to exert their anti-inflammatory effects due to: (1) inhibition of pro-inflammatory enzymes: quercetin and kaempferol inhibit the pro-inflammatory enzymes (cyclooxygenase and lipoxygenase); (2) regulation of cytokine production: isothiocyanates modulate signaling pathways involved in inflammation, such as the nuclear factor-kappa B (NF-kappa B) pathway; isothiocyanates inhibit the production of pro-inflammatory cytokines such as TNF-α (tumor necrosis factor α) and IL-1β (interleukin-1β); and (3) antioxidant activity: M. oleifera contains flavonoids, polyphenols, known to reduce oxidative stress and inflammation. The review includes M. oleifera's effects on cardiovascular protection, anti-hypertensive activities, type 2 diabetes, inflammatory bowel disease, and non-alcoholic fatty liver disease (NAFLD). This research could prove valuable for exploring the pharmacological potential of M. oleifera and contributing to the prospects of developing effective medicines for the benefit of human health.
Collapse
Affiliation(s)
- Adina Chiș
- Department of Molecular Sciences, “Iuliu Hațieganu” University of Medicine and Pharmacy, 6 Louis Pasteur St, 400349 Cluj-Napoca, Romania; (A.C.); (R.V.)
| | - Paul Aimé Noubissi
- Department of Animal Biology and Conservation, Faculty of Science, University of Buea, Buea P.O. Box 63, Cameroon; (P.A.N.); (R.K.)
| | - Oana-Lelia Pop
- Department of Food Science, University of Agricultural Science and Veterinary Medicine, 3-5 Calea Mănăștur, 400372 Cluj-Napoca, Romania; (O.-L.P.); (R.S.)
- Molecular Nutrition and Proteomics Lab, CDS3, Life Science Institute, University of Agricultural Science and Veterinary Medicine, 3-5 Calea Mănăștur, 400372 Cluj-Napoca, Romania
| | - Carmen Ioana Mureșan
- Department of Food Science, University of Agricultural Science and Veterinary Medicine, 3-5 Calea Mănăștur, 400372 Cluj-Napoca, Romania; (O.-L.P.); (R.S.)
- Molecular Nutrition and Proteomics Lab, CDS3, Life Science Institute, University of Agricultural Science and Veterinary Medicine, 3-5 Calea Mănăștur, 400372 Cluj-Napoca, Romania
| | - Michel Archange Fokam Tagne
- Department of Biological Sciences, Faculty of Science, University of Ngaoundéré, Ngaoundéré P.O. Box 454, Cameroon;
| | - René Kamgang
- Department of Animal Biology and Conservation, Faculty of Science, University of Buea, Buea P.O. Box 63, Cameroon; (P.A.N.); (R.K.)
| | - Adriana Fodor
- Clinical Center of Diabetes, Nutrition and Metabolic Diseases, “Iuliu Hațieganu” University of Medicine and Pharmacy, 2-4 Clinicilor St., 400012 Cluj-Napoca, Romania;
| | - Adela-Viviana Sitar-Tăut
- Department of Internal Medicine, Faculty of Medicine, “Iuliu Hațieganu” University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania; (A.-V.S.-T.); (A.C.); (O.H.O.)
| | - Angela Cozma
- Department of Internal Medicine, Faculty of Medicine, “Iuliu Hațieganu” University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania; (A.-V.S.-T.); (A.C.); (O.H.O.)
| | - Olga Hilda Orășan
- Department of Internal Medicine, Faculty of Medicine, “Iuliu Hațieganu” University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania; (A.-V.S.-T.); (A.C.); (O.H.O.)
| | - Simona Codruța Hegheș
- Department of Drug Analysis, Faculty of Pharmacy, “Iuliu Hațieganu” University of Medicine and Pharmacy, Louis Pasteur Street 6, 400349 Cluj-Napoca, Romania
| | - Romana Vulturar
- Department of Molecular Sciences, “Iuliu Hațieganu” University of Medicine and Pharmacy, 6 Louis Pasteur St, 400349 Cluj-Napoca, Romania; (A.C.); (R.V.)
| | - Ramona Suharoschi
- Department of Food Science, University of Agricultural Science and Veterinary Medicine, 3-5 Calea Mănăștur, 400372 Cluj-Napoca, Romania; (O.-L.P.); (R.S.)
- Molecular Nutrition and Proteomics Lab, CDS3, Life Science Institute, University of Agricultural Science and Veterinary Medicine, 3-5 Calea Mănăștur, 400372 Cluj-Napoca, Romania
| |
Collapse
|
12
|
Wang Z, Zhou Y, Xiao X, Liu A, Wang S, Preston RJS, Zaytseva YY, He G, Xiao W, Hennig B, Deng P. Inflammation and cardiometabolic diseases induced by persistent organic pollutants and nutritional interventions: Effects of multi-organ interactions. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2023; 339:122756. [PMID: 37844865 PMCID: PMC10842216 DOI: 10.1016/j.envpol.2023.122756] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/18/2023] [Revised: 10/11/2023] [Accepted: 10/13/2023] [Indexed: 10/18/2023]
Abstract
The development and outcome of inflammatory diseases are associated with genetic and lifestyle factors, which include chemical and nonchemical stressors. Persistent organic pollutants (POPs) are major groups of chemical stressors. For example, dioxin-like polychlorinated biphenyls (PCBs), per- and polyfluoroalkyl substances (PFASs), and polybrominated diphenyl ethers (PBDEs) are closely associated with the incidence of inflammatory diseases. The pathology of environmental chemical-mediated inflammatory diseases is complex and may involve disturbances in multiple organs, including the gut, liver, brain, vascular tissues, and immune systems. Recent studies suggested that diet-derived nutrients (e.g., phytochemicals, vitamins, unsaturated fatty acids, dietary fibers) could modulate environmental insults and affect disease development, progression, and outcome. In this article, mechanisms of environmental pollutant-induced inflammation and cardiometabolic diseases are reviewed, focusing on multi-organ interplays and highlighting recent advances in nutritional strategies to improve the outcome of cardiometabolic diseases associated with environmental exposures. In addition, advanced system biology approaches are discussed, which present unique opportunities to unveil the complex interactions among multiple organs and to fuel the development of precision intervention strategies in exposed individuals.
Collapse
Affiliation(s)
- Zhongmin Wang
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and College of Pharmaceutical Sciences, Soochow University, Suzhou, Jiangsu, China; Irish Centre for Vascular Biology, School of Pharmacy & Biomolecular Sciences, Royal College of Surgeons in Ireland, Ireland
| | - Yixuan Zhou
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and College of Pharmaceutical Sciences, Soochow University, Suzhou, Jiangsu, China
| | - Xia Xiao
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and College of Pharmaceutical Sciences, Soochow University, Suzhou, Jiangsu, China
| | - Aowen Liu
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and College of Pharmaceutical Sciences, Soochow University, Suzhou, Jiangsu, China
| | - Shengnan Wang
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and College of Pharmaceutical Sciences, Soochow University, Suzhou, Jiangsu, China
| | - Roger J S Preston
- Irish Centre for Vascular Biology, School of Pharmacy & Biomolecular Sciences, Royal College of Surgeons in Ireland, Ireland
| | - Yekaterina Y Zaytseva
- Superfund Research Center, University of Kentucky, Lexington, KY, USA; Department of Toxicology and Cancer Biology, University of Kentucky, Lexington, KY, USA
| | - Guangzhao He
- Department of Pharmacy, Changzhou Cancer Hospital, Soochow University, Changzhou, Jiangsu, China
| | - Wenjin Xiao
- Department of Endocrinology, Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Bernhard Hennig
- Superfund Research Center, University of Kentucky, Lexington, KY, USA; Department of Animal and Food Sciences, College of Agriculture, Food and Environment, University of Kentucky, Lexington, KY, USA
| | - Pan Deng
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and College of Pharmaceutical Sciences, Soochow University, Suzhou, Jiangsu, China.
| |
Collapse
|
13
|
Wang QC, Wang ZY. Comparative analysis of neutrophil-to-lymphocyte ratio and remnant cholesterol in predicting cardiovascular events and mortality in general adult population. Sci Rep 2023; 13:22362. [PMID: 38102174 PMCID: PMC10724289 DOI: 10.1038/s41598-023-49403-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Accepted: 12/07/2023] [Indexed: 12/17/2023] Open
Abstract
This study aimed to investigate the predictive value of neutrophil-to-lymphocyte ratio (NLR) and Remnant Cholesterol (Remnant-C) in relation to cardiovascular events and all-cause mortality in the general population. A population-based study. We conducted a retrospective cohort study analyzing data from the National Health and Nutrition Examination Survey (NHANES) spanning the years of 2011-2018, with follow-up for mortality status until December 31, 2019. Kaplan‒Meier and Cox proportional hazards regression analyses were used to evaluate the associations between NLR, Remnant-C, and cardiovascular events as well as all-cause mortality. Overall, 9409 individuals with both complete blood count and blood lipids were included in the analysis. Baseline NLR and Remnant-C were calculated. During the follow-up (median, 59.3 months), 177 cardiovascular events and 561 all-cause mortality occurred. In fully adjusted model, people with NLR > 2.26 were significantly associated with higher risk of cardiovascular events (HR 2.14, 95% CI 1.30-3.52, P < 0.001) and all-cause mortality (HR 1.66, 95% CI 1.30-2.12, P < 0.001). NLR exhibited a positive correlation with Remnant-C (r = 0.04, P < 0.001). Elevated NLR levels shown stronger association with cardiovascular events (HR 1.21, 95% CI 1.14-2.28, P < 0.001) compared with Remnant-C (HR 1.02, 95% CI 1.00-1.04, P = 0.020). Our findings suggest that NLR and Remnant-C are potential predictive markers for cardiovascular events in the general population. We observed a correlation between NLR and Remnant-C, and high NLR levels demonstrate a stronger association with the prediction of cardiovascular events and all-cause mortality compared with Remnant-C.
Collapse
Affiliation(s)
- Qian-Chen Wang
- Department of Cardiovascular Medicine, Xiangya Hospital, Central South University, Changsha, China
| | - Zhen-Yu Wang
- Department of Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, Renmin Road 139, Changsha, 410011, Hunan, China.
| |
Collapse
|
14
|
Gilly A, Park YC, Tsafantakis E, Karaleftheri M, Dedoussis G, Zeggini E. Genome-wide meta-analysis of 92 cardiometabolic protein serum levels. Mol Metab 2023; 78:101810. [PMID: 37778719 PMCID: PMC10582065 DOI: 10.1016/j.molmet.2023.101810] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Revised: 09/11/2023] [Accepted: 09/19/2023] [Indexed: 10/03/2023] Open
Abstract
OBJECTIVES Global cardiometabolic disease prevalence has grown rapidly over the years, making it the leading cause of death worldwide. Proteins are crucial components in biological pathways dysregulated in disease states. Identifying genetic components that influence circulating protein levels may lead to the discovery of biomarkers for early stages of disease or offer opportunities as therapeutic targets. METHODS Here, we carry out a genome-wide association study (GWAS) utilising whole genome sequencing data in 3,005 individuals from the HELIC founder populations cohort, across 92 proteins of cardiometabolic relevance. RESULTS We report 322 protein quantitative trait loci (pQTL) signals across 92 proteins, of which 76 are located in or near the coding gene (cis-pQTL). We link those association signals with changes in protein expression and cardiometabolic disease risk using colocalisation and Mendelian randomisation (MR) analyses. CONCLUSIONS The majority of previously unknown signals we describe point to proteins or protein interactions involved in inflammation and immune response, providing genetic evidence for the contributing role of inflammation in cardiometabolic disease processes.
Collapse
Affiliation(s)
- Arthur Gilly
- Institute of Translational Genomics, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, Germany
| | - Young-Chan Park
- Institute of Translational Genomics, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, Germany
| | | | | | - George Dedoussis
- Department of Nutrition and Dietetics, School of Health Science and Education, Harokopio University of Athens, Athens, Greece
| | - Eleftheria Zeggini
- Institute of Translational Genomics, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, Germany; Technical University of Munich (TUM) and Klinikum Rechts der Isar, TUM School of Medicine, Munich, Germany.
| |
Collapse
|
15
|
Cai Y, Yu R, Zhang Z, Li D, Yi B, Feng Z, Xu Q. Mettl3/Ythdf2 regulate macrophage inflammation and ROS generation by controlling Pyk2 mRNA stability. Immunol Lett 2023; 264:64-73. [PMID: 37952687 DOI: 10.1016/j.imlet.2023.11.004] [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: 04/13/2023] [Revised: 10/07/2023] [Accepted: 11/09/2023] [Indexed: 11/14/2023]
Abstract
As one of the most prevalent modifications on RNA, N6-methyladenosine (m6A) has been recently found implicated in various pathological processes. Emerging studies have demonstrated the role of m6A and its writer Mettl3 in fine-tuning the immune response, which now becomes a research hotspot owing to its potential therapeutic value. However, the results are inconsistent and even contradictory, suggesting that there might be multiple Mettl3 target genes involved in different pathways. To delve deeper into the function of Mettl3 in the cellular inflammatory response, we first conducted bioinformatics analysis using RNA-seq in Mettl3 ablation macrophages, and found that Mettl3 might attenuate LPS-induced proinflammatory pathways and reactive oxygen species (ROS) generation process. Mettl3 knockdown significantly increased the LPS-induced IL-6, TNF-α, NOXs (Nox1, Nox2, Ncf1, and Ncf2) expression, ROS generation, and the phosphorylation of MAPKs and AKT signaling. Combining the results of RNA-seq and m6A mapping, we found that Pyk2 might be the target gene of Mettl3 affecting the inflammatory response. Mettl3 and Ythdf2 depletion increased the expression and mRNA stability of Pyk2, and RIP-PCR showed that Ythdf2 directly targeting Pyk2 was Mettl3 dependent. Moreover, the upregulated expression of TNF-α, IL-6, NOXs, ROS generation, and the phosphorylation of MAPKs and AKT signaling were downregulated by Pyk2 inhibitor in Mettl3 knockdown cells. All of these results suggest that Mettl3 regulates the mRNA stability and expression of Pyk2 in a Ythdf2-dependent way, which consequently triggers the activation of MAPKs and AKT signaling and upregulation of NOXs, thus promoting the generation of proinflammatory cytokines and ROS.
Collapse
Affiliation(s)
- Yongjie Cai
- Hospital of Stomatology, Guanghua School of Stomatology, Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou 510055, China
| | - Ruiqing Yu
- Hospital of Stomatology, Guanghua School of Stomatology, Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou 510055, China
| | - Zhanqi Zhang
- Hospital of Stomatology, Guanghua School of Stomatology, Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou 510055, China
| | - Di Li
- Hospital of Stomatology, Guanghua School of Stomatology, Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou 510055, China
| | - Baicheng Yi
- Department of Stomatology, Shenzhen University General Hospital, Shenzhen University Clinical Medical Academy, Shenzhen 518055, China
| | - Zhihui Feng
- Hospital of Stomatology, Guanghua School of Stomatology, Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou 510055, China.
| | - Qiong Xu
- Hospital of Stomatology, Guanghua School of Stomatology, Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou 510055, China.
| |
Collapse
|
16
|
Florida EM, Li H, Hong CG, Ongstad EL, Gaddipati R, Sitaula S, Varma V, Parel PM, O'Hagan R, Chen MY, Teague HL, Playford MP, Karathanasis SK, Collén A, Mehta NN, Remaley AT, Sorokin AV. Relationship of Soluble Lectin-Like Low-Density Lipoprotein Receptor-1 (sLOX-1) With Inflammation and Coronary Plaque Progression in Psoriasis. J Am Heart Assoc 2023; 12:e031227. [PMID: 37982276 PMCID: PMC10727277 DOI: 10.1161/jaha.123.031227] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Accepted: 10/24/2023] [Indexed: 11/21/2023]
Abstract
BACKGROUND Psoriasis is a chronic inflammatory condition associated with coronary artery disease risk. Uptake of oxidized low-density lipoprotein by the lectin-like low-density lipoprotein receptor-1 triggers release of the soluble extracellular domain of the receptor (sLOX-1). We sought to characterize the relationship between sLOX-1, inflammation, and coronary plaque progression in psoriasis. METHODS AND RESULTS A total of 327 patients with psoriasis had serum sLOX-1 levels measured at baseline by an ELISA-based assay. Stratification by high-sensitivity C-reactive protein ≥4.0 mg/L (quartile 4), identified 81 participants who had coronary plaque phenotyping at baseline and were followed longitudinally by coronary computed tomography angiography. Subjects within high-sensitivity C-reactive protein quartile 4 were middle-aged (51.47±12.62 years), predominantly men (54.3%) with moderate psoriasis disease severity (6.60 [interquartile range, 3.30-13.40]). In the study cohort, participants with sLOX-1 above the median displayed increased vulnerable coronary plaque features. At baseline, sLOX-1 was associated with total burden (rho=0.296; P=0.01), noncalcified burden (rho=0.286; P=0.02), fibro-fatty burden (rho=0.346; P=0.004), and necrotic burden (rho=0.394; P=0.002). A strong relationship between sLOX-1, noncalcified burden (β=0.19; P=0.03), and fibro-fatty burden (β=0.29; P=0.003) was found in fully adjusted models at baseline and 1- and 4-year follow-up. Finally, coronary plaque features progressed over 1 year regardless of biologic or systemic treatment in subjects with high sLOX-1. CONCLUSIONS Patients with psoriasis with both high sLOX-1 and high-sensitivity C-reactive protein levels have increased coronary plaque burden associated with atherosclerotic plaque progression independent of biologic and systemic treatment. Thus, sLOX-1 might be considered as a promising marker in coronary artery disease risk estimation beyond traditional risk factors. REGISTRATION URL: https://www.clinicaltrials.gov; Unique identifier: NCT01778569.
Collapse
Affiliation(s)
- Elizabeth M. Florida
- Section of Inflammation and Cardiometabolic DiseasesNational Heart, Lung, and Blood Institute, National Institutes of HealthBethesdaMDUSA
| | - Haiou Li
- Section of Inflammation and Cardiometabolic DiseasesNational Heart, Lung, and Blood Institute, National Institutes of HealthBethesdaMDUSA
| | - Christin G. Hong
- Section of Inflammation and Cardiometabolic DiseasesNational Heart, Lung, and Blood Institute, National Institutes of HealthBethesdaMDUSA
| | - Emily L. Ongstad
- Bioscience Cardiovascular, Research and Early Development, Cardiovascular, Renal, and Metabolism (CVRM), BioPharmaceuticals R&D, AstraZenecaGaithersburgMDUSA
| | - Ranjitha Gaddipati
- Bioscience Cardiovascular, Research and Early Development, Cardiovascular, Renal, and Metabolism (CVRM), BioPharmaceuticals R&D, AstraZenecaGaithersburgMDUSA
| | - Sadichha Sitaula
- Bioscience Cardiovascular, Research and Early Development, Cardiovascular, Renal, and Metabolism (CVRM), BioPharmaceuticals R&D, AstraZenecaGaithersburgMDUSA
| | - Vijayalakshmi Varma
- Translational Science and Experimental Medicine, Research and Early Development, Cardiovascular, Renal, and Metabolism (CVRM), BioPharmaceuticals R&D, AstraZenecaGaithersburgMDUSA
| | - Philip M. Parel
- Section of Inflammation and Cardiometabolic DiseasesNational Heart, Lung, and Blood Institute, National Institutes of HealthBethesdaMDUSA
| | - Ross O'Hagan
- Section of Inflammation and Cardiometabolic DiseasesNational Heart, Lung, and Blood Institute, National Institutes of HealthBethesdaMDUSA
| | - Marcus Y. Chen
- Section of Inflammation and Cardiometabolic DiseasesNational Heart, Lung, and Blood Institute, National Institutes of HealthBethesdaMDUSA
| | - Heather L. Teague
- Section of Inflammation and Cardiometabolic DiseasesNational Heart, Lung, and Blood Institute, National Institutes of HealthBethesdaMDUSA
| | - Martin P. Playford
- Section of Inflammation and Cardiometabolic DiseasesNational Heart, Lung, and Blood Institute, National Institutes of HealthBethesdaMDUSA
| | - Sotirios K. Karathanasis
- NeoProgenBaltimoreMDUSA
- Section of Lipoprotein Metabolism, Translational Vascular Medicine Branch, National HeartLung and Blood Institute, National Institutes of HealthBethesdaMDUSA
| | - Anna Collén
- Projects, Research and Early Development, Cardiovascular, Renal, and Metabolism (CVRM), BioPharmaceuticals R&D, AstraZenecaGaithersburgMDUSA
| | - Nehal N. Mehta
- Section of Inflammation and Cardiometabolic DiseasesNational Heart, Lung, and Blood Institute, National Institutes of HealthBethesdaMDUSA
| | - Alan T. Remaley
- Section of Lipoprotein Metabolism, Translational Vascular Medicine Branch, National HeartLung and Blood Institute, National Institutes of HealthBethesdaMDUSA
| | - Alexander V. Sorokin
- Section of Inflammation and Cardiometabolic DiseasesNational Heart, Lung, and Blood Institute, National Institutes of HealthBethesdaMDUSA
| |
Collapse
|
17
|
Oh YJ, Jin SE, Shin HK, Ha H. Daeshiho-tang attenuates inflammatory response and oxidative stress in LPS-stimulated macrophages by regulating TLR4/MyD88, NF-κB, MAPK, and Nrf2/HO-1 pathways. Sci Rep 2023; 13:18891. [PMID: 37919391 PMCID: PMC10622541 DOI: 10.1038/s41598-023-46033-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Accepted: 10/26/2023] [Indexed: 11/04/2023] Open
Abstract
Daeshiho-tang (DSHT), a traditional herbal formula with diverse pharmacological effects, has shown promise in medicine owing to its anti-hypertensive, anti-diabetic, and anti-inflammatory properties. However, the precise molecular mechanism underlying these effects remains unclear. Thus, we investigated the effect of DSHT on inflammatory response and oxidative stress to understand its molecular mechanism using lipopolysaccharide (LPS)-induced macrophage (RAW 264.7) cells. DSHT decreased the contents of nitric oxide (NO) and prostaglandin E2 (PGE2) through downregulating inducible nitric oxide synthase (iNOS) and cyclooxygenase-2 (COX-2) protein expressions. DSHT suppressed the LPS-induced TLR4 as well as MyD88, subsequently suppressing the NF-κB activation and the phosphorylation of MAPK (p38, ERK, and JNK). Radical scavenging activity results revealed a dose-dependent response of DSHT with diminished ABTS activity, a hallmark of oxidative stress potential. Furthermore, DSHT enhanced Nrf2 and HO-1 expression in response to LPS. Collectively, our findings indicated that DSHT exert anti-inflammatory effect and regulating oxidative stress by modulating TLR4/MyD88, NF-κB, MAPK, and Nrf2/HO-1 pathways, consequently can provide potential therapeutic strategy for the prevention and treatment of inflammation and oxidative stress-related diseases.
Collapse
Affiliation(s)
- Yong Jin Oh
- KM Science Research Division, Korea Institute of Oriental Medicine, 1672 Yuseong-daero, Yuseong-Gu, Daejeon, 34054, Korea
| | - Seong Eun Jin
- KM Science Research Division, Korea Institute of Oriental Medicine, 1672 Yuseong-daero, Yuseong-Gu, Daejeon, 34054, Korea
| | - Hyeun-Kyoo Shin
- KM Science Research Division, Korea Institute of Oriental Medicine, 1672 Yuseong-daero, Yuseong-Gu, Daejeon, 34054, Korea
| | - Hyekyung Ha
- KM Science Research Division, Korea Institute of Oriental Medicine, 1672 Yuseong-daero, Yuseong-Gu, Daejeon, 34054, Korea.
| |
Collapse
|
18
|
Johnson CM, Talluru SM, Bubic B, Colbert M, Kumar P, Tsai HL, Varadhan R, Rozati S. Association of Cardiovascular Disease in Patients with Mycosis Fungoides and Sézary Syndrome Compared to a Matched Control Cohort. JID INNOVATIONS 2023; 3:100219. [PMID: 38116332 PMCID: PMC10730311 DOI: 10.1016/j.xjidi.2023.100219] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Revised: 06/02/2023] [Accepted: 06/07/2023] [Indexed: 12/21/2023] Open
Abstract
Mycosis fungoides/Sézary syndrome (MF/SS) produces a low-grade chronic inflammatory state that may be associated with an increased risk of cardiovascular (CV) events, as seen in other chronic, systemic dermatologic diseases. To assess this association, a retrospective, cross-sectional study was designed in which 421 patients with a biopsy-proven diagnosis of MF/SS were compared with a control cohort of 4,210 age-, gender-, and race-matched patients randomly selected from the National Health and Nutritional Evaluation Survey database. The MF/SS cohort had a 14% prevalence of CV events, which was not statistically different from the control population's prevalence of 13%. In the MF/SS cohort, a multivariable logistic regression model showed that older patients (OR = 1.05 for each year of age, 95% confidence interval = 1.02-1.07) and those diagnosed with hypertension (OR = 3.40, 95% confidence interval = 1.71-6.75) had a higher risk of a CV event (P < 0.001). Risk factors such as gender, race, smoking, diabetes, and obesity were not significantly associated with CV events. Findings suggest that in the MF/SS population, advancing age and hypertension are risk factors for CV events, requiring clinical recognition and management. In addition, further research is needed to understand the complex interplay of how chronic inflammation in MF/SS impacts the immune development of CV disease.
Collapse
Affiliation(s)
- Courtney M Johnson
- Department of Dermatology, Johns Hopkins School of Medicine, Baltimore, Maryland
| | - Sai M Talluru
- Department of Dermatology, Johns Hopkins School of Medicine, Baltimore, Maryland
| | - Bianka Bubic
- Department of Dermatology, Johns Hopkins School of Medicine, Baltimore, Maryland
| | - Michelle Colbert
- Department of Dermatology, Johns Hopkins School of Medicine, Baltimore, Maryland
| | - Priyanka Kumar
- Department of Dermatology, Johns Hopkins School of Medicine, Baltimore, Maryland
| | - Hua-Ling Tsai
- Quantitative Sciences Division, Department of Oncology, Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| | - Ravi Varadhan
- Quantitative Sciences Division, Department of Oncology, Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| | - Sima Rozati
- Department of Dermatology, Johns Hopkins School of Medicine, Baltimore, Maryland
| |
Collapse
|
19
|
Cho S, Ying F, Sweeney G. Sterile inflammation and the NLRP3 inflammasome in cardiometabolic disease. Biomed J 2023; 46:100624. [PMID: 37336361 PMCID: PMC10539878 DOI: 10.1016/j.bj.2023.100624] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Revised: 06/11/2023] [Accepted: 06/14/2023] [Indexed: 06/21/2023] Open
Abstract
Inflammation plays an important role in the pathophysiology of cardiometabolic diseases. Sterile inflammation, a non-infectious and damage-associated molecular pattern (DAMP)-induced innate response, is now well-established to be closely associated with development and progression of cardiometabolic diseases. The NOD-like receptor (NLR) family pyrin domain-containing 3 (NLRP3) inflammasome is well-established as a major player in sterile inflammatory responses. It is a multimeric cytosolic protein complex which regulates the activation of caspase-1 and subsequently promotes cleavage and release of interleukin (IL)-1 family cytokines, which have a deleterious impact on the development of cardiometabolic diseases. Therefore, targeting NLRP3 itself or the downstream consequences of NLRP3 activation represent excellent potential therapeutic targets in inflammatory cardiometabolic diseases. Here, we review our current understanding of the role which NLRP3 inflammasome regulation plays in cardiometabolic diseases such as obesity, diabetes, non-alcoholic steatohepatitis (NASH), atherosclerosis, ischemic heart disease and cardiomyopathy. Finally, we highlight the potential of targeting NLPR3 or related signaling molecules as a therapeutic approach.
Collapse
Affiliation(s)
- Sungji Cho
- Department of Biology, York University, Toronto, Ontario, Canada
| | - Fan Ying
- Department of Biology, York University, Toronto, Ontario, Canada
| | - Gary Sweeney
- Department of Biology, York University, Toronto, Ontario, Canada.
| |
Collapse
|
20
|
O'Hagan R, Hsu LY, Li H, Hong CG, Parel PM, Berg AR, Manyak GA, Bui V, Patel NH, Florida EM, Teague HL, Playford MP, Zhou W, Dey D, Chen MY, Mehta NN, Sorokin AV. Longitudinal association of epicardial and thoracic adipose tissues with coronary and cardiac characteristics in psoriasis. Heliyon 2023; 9:e20732. [PMID: 37867905 PMCID: PMC10585224 DOI: 10.1016/j.heliyon.2023.e20732] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Revised: 10/03/2023] [Accepted: 10/05/2023] [Indexed: 10/24/2023] Open
Abstract
Background s: Psoriasis is a disease of systemic inflammation associated with increased cardiometabolic risk. Epicardial adipose tissue (EAT) and thoracic adipose tissue (TAT) are contributing factors for atherosclerosis and cardiac dysfunction. We strove to assess the longitudinal impact of the EAT and TAT on coronary and cardiac characteristics in psoriasis. Methods The study consisted of 301 patients with baseline coronary computed tomography angiography (CTA), of which 139 had four-year follow up scans. EAT and TAT volumes from non-contrast computed tomography scans were quantified by an automated segmentation framework. Coronary plaque characteristics and left ventricular (LV) mass were quantified by CTA. Results When stratified by baseline EAT and TAT volume quartiles, a stepwise significant increase in cardiometabolic parameters was observed. EAT and TAT volumes associated with fibro-fatty burden (FFB) (TAT: ρ = 0.394, P < 0.001; EAT: ρ = 0.459, P < 0.001) in adjusted models. Only EAT had a significant four-year time-dependent association with FFB in fully adjusted models (β = 0.307 P = 0.003), whereas only TAT volume associated with myocardial injury in fully adjusted models (TAT: OR = 1.57 95 % CI = (1.00-2.60); EAT: OR = 1.46 95 % CI = (0.91-2.45). Higher quartiles of EAT and TAT had increased LV mass and developed strong correlation (TAT: ρ = 0.370, P < 0.001; EAT: ρ = 0.512, P < 0.001). Conclusions Our study is the first to explore how both EAT and TAT volumes associate with increased cardiometabolic risk profile in an inflamed psoriasis cohorts and highlight the need for further studies on its use as a potential prognostic tool for high-risk coronary plaques and cardiac dysfunction.
Collapse
Affiliation(s)
- Ross O'Hagan
- Section of Inflammation and Cardiometabolic Diseases, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Li-Yueh Hsu
- Radiology and Imaging Sciences, Clinical Center, National Institutes of Health, Bethesda, MD, USA
| | - Haiou Li
- Section of Inflammation and Cardiometabolic Diseases, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Christin G. Hong
- Section of Inflammation and Cardiometabolic Diseases, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Philip M. Parel
- Section of Inflammation and Cardiometabolic Diseases, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Alexander R. Berg
- Section of Inflammation and Cardiometabolic Diseases, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Grigory A. Manyak
- Section of Inflammation and Cardiometabolic Diseases, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Vy Bui
- Radiology and Imaging Sciences, Clinical Center, National Institutes of Health, Bethesda, MD, USA
| | - Nidhi H. Patel
- Section of Inflammation and Cardiometabolic Diseases, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Elizabeth M. Florida
- Section of Inflammation and Cardiometabolic Diseases, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Heather L. Teague
- Section of Inflammation and Cardiometabolic Diseases, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Martin P. Playford
- Section of Inflammation and Cardiometabolic Diseases, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Wunan Zhou
- Section of Inflammation and Cardiometabolic Diseases, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Damini Dey
- Biomedical Imaging Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Marcus Y. Chen
- Section of Inflammation and Cardiometabolic Diseases, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Nehal N. Mehta
- Section of Inflammation and Cardiometabolic Diseases, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Alexander V. Sorokin
- Section of Inflammation and Cardiometabolic Diseases, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
21
|
Hong CG, Florida E, Li H, Parel PM, Mehta NN, Sorokin AV. Oxidized low-density lipoprotein associates with cardiovascular disease by a vicious cycle of atherosclerosis and inflammation: A systematic review and meta-analysis. Front Cardiovasc Med 2023; 9:1023651. [PMID: 36727024 PMCID: PMC9885196 DOI: 10.3389/fcvm.2022.1023651] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2022] [Accepted: 12/30/2022] [Indexed: 01/18/2023] Open
Abstract
Background Low-density lipoprotein cholesterol (LDL-C) is an established marker for cardiovascular disease (CVD) and a therapeutic target. Oxidized LDL (oxLDL) is known to be associated with excessive inflammation and abnormal lipoprotein metabolism. Chronic inflammatory diseases confer an elevated risk of premature atherosclerosis and adverse cardiovascular events. Whether oxLDL may serve as a potential biomarker for CVD stratification in populations with chronic inflammatory conditions remains understudied. Objective To perform a systematic review and meta-analysis evaluating the relationship between oxLDL and CVD (defined by incident CVD events, carotid intima-media thickness, presence of coronary plaque) in patients with chronic inflammatory diseases. Methods A systematic literature search was performed using studies published between 2000 and 2022 from PubMed, Cochrane Library, Embase (Elsevier), CINHAL (EBSCOhost), Scopus (Elsevier), and Web of Science: Core Collection (Clarivate Analytics) databases on the relationship between oxLDL and cardiovascular risk on inflamed population. The pooled effect size was combined using the random effect model and publication bias was assessed if P < 0.05 for the Egger or Begg test along with the funnel plot test. Results A total of three observational studies with 1,060 participants were ultimately included in the final meta-analysis. The results demonstrated that oxLDL is significantly increased in participants with CVD in the setting of chronic inflammatory conditions. This meta-analysis suggests that oxLDL may be a useful biomarker in risk stratifying cardiovascular disease in chronically inflamed patients.
Collapse
|
22
|
Delbaere Q, Chapet N, Huet F, Delmas C, Mewton N, Prunier F, Angoulvant D, Roubille F. Anti-Inflammatory Drug Candidates for Prevention and Treatment of Cardiovascular Diseases. Pharmaceuticals (Basel) 2023; 16:78. [PMID: 36678575 PMCID: PMC9865197 DOI: 10.3390/ph16010078] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2022] [Revised: 12/26/2022] [Accepted: 12/28/2022] [Indexed: 01/07/2023] Open
Abstract
Incidence and mortality rates for cardiovascular disease are declining, but it still remains a major cause of morbidity and mortality. Drug treatments to slow the progression of atherosclerosis focus on reducing cholesterol levels. The paradigm shift to consider atherosclerosis an inflammatory disease by itself has led to the development of new treatments. In this article, we discuss the pathophysiology of inflammation and focus attention on therapeutics targeting different inflammatory pathways of atherosclerosis and myocardial infarction. In atherosclerosis, colchicine is included in new recommendations, and eight randomized clinical trials are testing new drugs in different inflammatory pathways. After a myocardial infarction, no drug has shown a significant benefit, but we present four randomized clinical trials with new treatments targeting inflammation.
Collapse
Affiliation(s)
- Quentin Delbaere
- Department of Cardiology, Arnaud de Villeneuve University Hospital, 34295 Montpellier, France
| | - Nicolas Chapet
- Department of Cardiology, Arnaud de Villeneuve University Hospital, 34295 Montpellier, France
| | - Fabien Huet
- Department of Cardiology, Arnaud de Villeneuve University Hospital, 34295 Montpellier, France
- Department of Cardiology, Bretagne Atlantique General Hospital, 56000 Vannes, France
| | - Clément Delmas
- Department of Cardiology, Arnaud de Villeneuve University Hospital, 34295 Montpellier, France
| | - Nathan Mewton
- Hôpital Cardiovasculaire Louis Pradel, 69002 Lyon, France
| | - Fabrice Prunier
- Department of Cardiology, CHU Angers, Université d’Angers, 49100 Angers, France
| | - Denis Angoulvant
- Cardiology Department, CHRU de Tours, 37044 Tours, France
- EA 4245 T2I, Université de Tours, 37044 Tours, France
| | - François Roubille
- Department of Cardiology, Arnaud de Villeneuve University Hospital, 34295 Montpellier, France
| |
Collapse
|
23
|
Liu S, Liu F, Zhang Z, Zhuang Z, Yuan X, Chen Y. The SELP, CD93, IL2RG, and VAV1 Genes Associated with Atherosclerosis May Be Potential Diagnostic Biomarkers for Psoriasis. J Inflamm Res 2023; 16:827-843. [PMID: 36876153 PMCID: PMC9983575 DOI: 10.2147/jir.s398862] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Accepted: 02/16/2023] [Indexed: 03/03/2023] Open
Abstract
Purpose Psoriasis and atherosclerosis are immunometabolic diseases. This study aimed to integrate bioinformatics and updated public resources to find potential biological markers associated with atherosclerosis that can cause psoriasis. Patients and Methods Microarray datasets were downloaded from the Gene Expression Omnibus (GEO) database. Differentially expressed genes (DEGs) were screened, and functional enrichment analysis was performed. We identified psoriasis and atherosclerosis common immune-related genes (PA-IRGs) by overlapping immune-related genes (IRGs) with genes in the module most associated with psoriasis and atherosclerosis obtained by weighted gene co-expression network analysis (WGCNAs). Receiver operating characteristic (ROC) was conducted to evaluate the predictive ability. The skin expression levels of diagnostic biomarkers were further verified by immunohistochemical staining. CIBERSORT, single-sample gene set enrichment analysis (ssGSEA), and Pearson's correlation analysis were applied to evaluate immune and lipid metabolism relationships in psoriatic tissues. In addition, a lincRNA-miRNA-mRNA network was constructed to find the pathogenesis in which diagnostic markers may be involved. Results Four PA-IRGs (SELP, CD93, IL2RG, and VAV1) demonstrated the optimal diagnostic value, with an AUC above 0.8. The immune cell infiltration analysis showed that dendritic resting cells, NK cell activation, neutrophils, macrophages M2, macrophages M0, and B-cell memory were highly abundant in psoriasis. Immune response analysis showed that TNF family members, chemokine receptors, interferons, natural killer cells, and TGF-β family members might be involved in psoriasis. Diagnostic biomarkers are strongly associated with various infiltrating immune cells, immune responses, and lipid metabolism. A lincRNA-miRNA-mRNA regulatory network consisting of 31 lincRNAs and 23 miRNAs was constructed. LINC00662 is involved in modulating four diagnostic biomarkers. Conclusion This study identified atherosclerosis-related genes SELP, CD93, VAV1, and IL2RG as potential psoriasis diagnostic markers. Provide novel insights into the possible regulatory mechanisms involved in psoriasis.
Collapse
Affiliation(s)
- Shougang Liu
- Department of Dermatology, Dermatology Hospital of Southern Medical University, Guangzhou, People's Republic of China
| | - Fanghua Liu
- Department of Dermatology, Dermatology Hospital of Southern Medical University, Guangzhou, People's Republic of China.,Department of Dermatology, Guangdong Medical University, Zhanjiang, People's Republic of China
| | - Zeqiao Zhang
- Department of Dermatology, Dermatology Hospital of Southern Medical University, Guangzhou, People's Republic of China
| | - Zhe Zhuang
- Department of Dermatology, Dermatology Hospital of Southern Medical University, Guangzhou, People's Republic of China.,Department of Dermatology, Guangdong Medical University, Zhanjiang, People's Republic of China
| | - Xiuqing Yuan
- Department of Dermatology, Dermatology Hospital of Southern Medical University, Guangzhou, People's Republic of China
| | - Yongfeng Chen
- Department of Dermatology, Dermatology Hospital of Southern Medical University, Guangzhou, People's Republic of China.,Department of Dermatology, Guangdong Medical University, Zhanjiang, People's Republic of China
| |
Collapse
|
24
|
Zhu C, Ren Y, Yao H, Feng B, Liu L, Zheng M. Heparanase Contributes to Psoriatic Lesions Through Crosstalk with IL-17 Pathway. Indian J Dermatol 2023; 68:59-66. [PMID: 37151254 PMCID: PMC10162766 DOI: 10.4103/ijd.ijd_641_22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/09/2023] Open
Abstract
Psoriasis is a chronic inflammatory disease that is considered by a network of immunocytes and cytokines. Among all, Th17 cells-derived IL-17 is a critical driving factor in the pathogenesis of psoriasis. Recently, disruption of the extracellular matrix was found to be related to psoriasis progression. In the present study, we aimed to investigate the role of heparanase (HPSE) in psoriasis and the crosstalk with the IL-17 signalling pathway. Skin tissues from non-affected areas and psoriatic lesion areas before and after 12 weeks of IL-17 monoclonal antibody treatment of 30 psoriasis patients were collected. HaCaT cells were treated with different concentrations of IL-17 antibody, and HPSE in cells and medium were measured with Western blotting assay as well as enzyme-linked immunosorbent assay (ELISA). In the imiquimod (IMQ)-induced psoriasis model, IL-17 protein and mRNA expression levels were measured, and changes in the proportion of Th17 cells were detected via flow cytometry. Our data showed that HPSE is upregulated in lesion tissues isolated from psoriasis patients, and was inhibited by anti-IL-17 treatment. In cutaneous cells and IMQ-induced psoriasis model, IL-17 promoted the synthesis of HPSE. Inversely, HPSE was also found to increase the percentage of Th17 cells derived from CD4+ T cells. Finally, we found that the combined treatments of HPSE inhibitor and IL-17 monoclonal antibody produced therapeutic effects on IMQ-induced psoriasis model. Our findings revealed the new role of HPSE in the pathogenesis of psoriasis and also provided a target for combined treatment of psoriasis.
Collapse
Affiliation(s)
- Chengyao Zhu
- From the Department of Dermatology, The Fourth Affiliated Hospital of Zhejiang University School of Medicine, Yiwu, China
- Department of Dermatology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Yunqin Ren
- Department of Dermatology, The Children's Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Hongliang Yao
- From the Department of Dermatology, The Fourth Affiliated Hospital of Zhejiang University School of Medicine, Yiwu, China
| | - Bo Feng
- From the Department of Dermatology, The Fourth Affiliated Hospital of Zhejiang University School of Medicine, Yiwu, China
| | - Lunfei Liu
- From the Department of Dermatology, The Fourth Affiliated Hospital of Zhejiang University School of Medicine, Yiwu, China
| | - Min Zheng
- Department of Dermatology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
25
|
Glucogallin Attenuates the LPS-Induced Signaling in Macrophages and Protects Mice against Sepsis. Int J Mol Sci 2022; 23:ijms231911254. [PMID: 36232563 PMCID: PMC9569803 DOI: 10.3390/ijms231911254] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Revised: 09/06/2022] [Accepted: 09/13/2022] [Indexed: 11/17/2022] Open
Abstract
The anti-oxidant and anti-inflammatory effect of beta-glucogallin (BGG), a plant-derived natural product, was evaluated in both in vitro and in vivo studies. For the in vitro study, the ability of BGG pre-treatment to quench LPS-induced effects compared to LPS alone in macrophages was investigated. It was found that BGG pre-treatment showed a significant decrease in ROS, NO, superoxide, and pro-inflammatory cytokines (TNF-alpha, IL-4, IL-17, IL-1β, and IL-6) and increased reduced glutathione coupled with the restoration of mitochondrial membrane potential. Gene profiling and further validation by qPCR showed that BGG pre-treatment downregulated the LPS-induced expression of c-Fos, Fas, MMP-9, iNOS, COX-2, MyD88, TRIF, TRAF6, TRAM, c-JUN, and NF-κB. We observed that BGG pre-treatment reduced nuclear translocation of LPS-activated NF-κB and thus reduced the subsequent expressions of NLRP3 and IL-1β, indicating the ability of BGG to inhibit inflammasome formation. Molecular docking studies showed that BGG could bind at the active site of TLR4. Finally, in the LPS-driven sepsis mouse model, we showed that pre-treatment with BGG sustained toxic shock, as evident from their 100% survival. Our study clearly showed the therapeutic potential of BGG in toxic shock syndrome.
Collapse
|
26
|
Graceffa D, Sperati F, Bonifati C, Spoletini G, Lora V, Pimpinelli F, Pontone M, Pellini R, Di Bella O, Morrone A, Cristaudo A. Immunogenicity of three doses of anti-SARS-CoV-2 BNT162b2 vaccine in psoriasis patients treated with biologics. Front Med (Lausanne) 2022; 9:961904. [PMID: 36148445 PMCID: PMC9485492 DOI: 10.3389/fmed.2022.961904] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2022] [Accepted: 08/15/2022] [Indexed: 11/16/2022] Open
Abstract
INTRODUCTION Psoriasis has not been directly linked to a poor prognosis for COVID-19, yet immunomodulatory agents used for its management may lead to increased vulnerability to the dangerous complications of SARS-CoV-2 infection, as well as impair the effectiveness of the recently introduced vaccines. The three-dose antibody response trend and the safety of BNT162b2 mRNA vaccine in psoriasis patients treated with biologic drugs have remained under-researched. MATERIALS AND METHODS Forty-five psoriatic patients on biologic treatment were enrolled to evaluate their humoral response to three doses of BNT162b2. IgG titers anti-SARS-CoV-2 spike protein were evaluated at baseline (day 0, first dose), after 3 weeks (second dose), four weeks post-second dose, at the time of the third dose administration and 4 weeks post-third dose. Seropositivity was defined as IgG ≥15 antibody-binding units (BAU)/mL. Data on vaccine safety were also collected by interview at each visit. RESULTS A statistically significant increase in antibody titers was observed after each dose of vaccine compared with baseline, with no significant differences between patients and controls. Methotrexate used in combination with biologics has been shown to negatively influence the antibody response to the vaccine. On the contrary, increasing body mass index (BMI) positively influenced the antibody response. No adverse effects were reported, and no relapses of psoriasis were observed in the weeks following vaccine administration in our study population. CONCLUSIONS Our data are largely consistent with the recent literature on this topic confirming the substantial efficacy and safety of BNT162b2 mRNA vaccine on psoriatic patients treated with biologics of different types and support the recommendation to perform additional doses in this specific subgroup of patients.
Collapse
Affiliation(s)
- Dario Graceffa
- Department of Clinical Dermatology, San Gallicano Dermatological Institute, IRCCS, Rome, Italy
| | - Francesca Sperati
- Biostatistics Unit, San Gallicano Dermatological Institute IRCCS, Rome, Italy
| | - Claudio Bonifati
- Department of Clinical Dermatology, San Gallicano Dermatological Institute, IRCCS, Rome, Italy
| | - Gabriele Spoletini
- General Surgery and Liver Transplantation, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Viviana Lora
- Department of Clinical Dermatology, San Gallicano Dermatological Institute, IRCCS, Rome, Italy
| | - Fulvia Pimpinelli
- Microbiology and Virology Unit, Dermatological Clinical and Research Department, San Gallicano Dermatological Institute, IRCCS, Rome, Italy
| | - Martina Pontone
- Microbiology and Virology Unit, Dermatological Clinical and Research Department, San Gallicano Dermatological Institute, IRCCS, Rome, Italy
| | - Raul Pellini
- Department Otolaryngology Head and Neck Surgery, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Ornella Di Bella
- Medical Direction, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Aldo Morrone
- Scientific Direction, San Gallicano Dermatological Institute, IRCCS, Rome, Italy
| | - Antonio Cristaudo
- Department of Clinical Dermatology, San Gallicano Dermatological Institute, IRCCS, Rome, Italy
| |
Collapse
|
27
|
Kohsari M, Moradinazar M, Rahimi Z, Najafi F, Pasdar Y, Shakiba E. New inflammatory biomarkers (lymphocyte and monocyte percentage to high-density lipoprotein cholesterol ratio and lymphocyte to monocyte percentage ratio) and their association with some cardiometabolic diseases : Results from a large Kurdish cohort study in Iran. Wien Klin Wochenschr 2022; 134:626-635. [PMID: 35471616 DOI: 10.1007/s00508-022-02029-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Accepted: 03/17/2022] [Indexed: 12/30/2022]
Abstract
BACKGROUND The incidence of metabolic heart diseases has increased significantly in Middle Eastern countries such as Iran. The present study aimed to investigate the association between monocyte percentage to high-density cholesterol ratio (MHR), lymphocyte percentage to high-density cholesterol ratio (LHR), and lymphocyte to monocyte percentage ratio (LMR) and cardiometabolic diseases in a Kurdish population in the west of Iran. METHODS This study recruited 9803 individuals, 4728 (48.2%) were male and 5084 (51.8%) were female from Ravansar, Iran. All biomarkers were analyzed by the standard methods. RESULTS The prevalence of cardiometabolic diseases was higher in overweight/obese participants and increased with age. MHR and LHR increased significantly in cardiometabolic individuals compared with healthy controls. Individuals in the fourth quartiles of LHR and MHR had higher odds ratio (ORs) for metabolic syndrome (MetS) and diabetes mellitus (DM) than the first quartiles. The LMR had a statistical association with non-alcoholic fatty liver disease (NAFLD) ORs and FLI. Besides, all these associations were stronger for females, and increased physical activity decreased inflammatory biomarkers. CONCLUSION The present study showed MHR and LHR had significant associations with ORs of MetS and DM. Also, MHR and LHR had a significant positive correlation with cardiometabolic risk factors. The LMR only had a statistical association with NAFLD and fatty liver index (FLI). Besides, the strong correlation between inflammatory biomarkers and cardiometabolic risk factors in females might be relevant to higher fat accumulation and metabolic inflammation background, and lower physical activity.
Collapse
Affiliation(s)
- Maryam Kohsari
- Behavioral disease Research Center, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Mehdi Moradinazar
- Behavioral disease Research Center, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Zohreh Rahimi
- Behavioral disease Research Center, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Farid Najafi
- Behavioral disease Research Center, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Yahya Pasdar
- Behavioral disease Research Center, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Ebrahim Shakiba
- Behavioral disease Research Center, Kermanshah University of Medical Sciences, Kermanshah, Iran.
- School of Medicine, St. University, Shahid Shiroudi Boulevard, 6714869914, Kermanshah, Iran.
| |
Collapse
|
28
|
Li W, Luo F, Wu X, Fan B, Yang M, Zhong W, Guan D, Wang F, Wang Q. Anti-Inflammatory Effects and Mechanisms of Dandelion in RAW264.7 Macrophages and Zebrafish Larvae. Front Pharmacol 2022; 13:906927. [PMID: 36091818 PMCID: PMC9454954 DOI: 10.3389/fphar.2022.906927] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Accepted: 06/23/2022] [Indexed: 11/13/2022] Open
Abstract
Dandelions (Taraxacum spp.) play an important role in the treatment of inflammatory diseases. In this study, we investigated the anti-inflammatory effects of Dandelion Extract (DE) in LPS-induced RAW264.7 macrophages and copper sulfate (CuSO4)-induced zebrafish larvae. DE was not toxic to RAW264.7 cells at 75 μg/ml as measured by cell viability, and DE inhibited LPS-induced cell morphological changes as measured by inverted microscopy. In survival experiments, DE at 25 μg/ml had no toxicity to zebrafish larvae. By using an enzymatic standard assay, DE reduced the production of nitric oxide (NO) in LPS-induced RAW264.7 cells. Fluorescence microscopy results show that DE reduced LPS-induced ROS production and apoptosis in RAW264.7 cells. DE also inhibited CuSO4-induced ROS production and neutrophil aggregation in zebrafish larvae. The results of flow cytometry show that DE alleviated the LPS-induced cell cycle arrest. In LPS-induced RAW264.7 cells, RT-PCR revealed that DE decreased the expression of M1 phenotypic genes iNOS, IL-6, and IL-1β while increasing the expression of M2 phenotypic genes IL-10 and CD206. Furthermore, in CuSO4-induced zebrafish larvae, DE reduced the expression of iNOS, TNF-α, IL-6, and IL-10. The findings suggest that DE reduces the LPS-induced inflammatory response in RAW264.7 cells by regulating polarization and apoptosis. DE also reduces the CuSO4-induced inflammatory response in zebrafish larvae.
Collapse
Affiliation(s)
- Wenju Li
- Institute of Food Science and Technology, Chinese Academy of Agricultural Sciences, Beijing, China
- Department of Emergency Medicine, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Fulong Luo
- Institute of Food Science and Technology, Chinese Academy of Agricultural Sciences, Beijing, China
- Department of Emergency Medicine, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Xiaohui Wu
- Department of Neurology, The First Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Bei Fan
- Institute of Food Science and Technology, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Mingran Yang
- Institute of Food Science and Technology, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Wu Zhong
- Department of Emergency Medicine, The Affiliated Hospital of Southwest Medical University, Luzhou, China
- Sichuan Provincial Rehabilitation Hospital, Chengdu, China
| | - Dongyan Guan
- Institute of Food Science and Technology, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Fengzhong Wang
- Institute of Food Science and Technology, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Qiong Wang
- Institute of Food Science and Technology, Chinese Academy of Agricultural Sciences, Beijing, China
- Sino-Portugal TCM International Cooperation Center, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, China
| |
Collapse
|
29
|
Shi H, Wu H, Winkler MA, Belin de Chantemèle EJ, Lee R, Kim HW, Weintraub NL. Perivascular adipose tissue in autoimmune rheumatic diseases. Pharmacol Res 2022; 182:106354. [PMID: 35842184 PMCID: PMC10184774 DOI: 10.1016/j.phrs.2022.106354] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Revised: 06/27/2022] [Accepted: 07/11/2022] [Indexed: 01/14/2023]
Abstract
Perivascular adipose tissue (PVAT) resides at the outermost boundary of the vascular wall, surrounding most conduit blood vessels, except for the cerebral vessels, in humans. A growing body of evidence suggests that inflammation localized within PVAT may contribute to the pathogenesis of cardiovascular disease (CVD). Patients with autoimmune rheumatic diseases (ARDs), e.g., systemic lupus erythematosus (SLE), rheumatoid arthritis (RA), psoriasis, etc., exhibit heightened systemic inflammation and are at increased risk for CVD. Data from clinical studies in patients with ARDs support a linkage between dysfunctional adipose tissue, and PVAT in particular, in disease pathogenesis. Here, we review the data linking PVAT to the pathogenesis of CVD in patients with ARDs, focusing on the role of novel PVAT imaging techniques in defining disease risk and responses to biological therapies.
Collapse
Affiliation(s)
- Hong Shi
- Division of Rheumatology, Medical College of Georgia at Augusta University, Augusta, GA, USA; Vascular Biology Center, Medical College of Georgia at Augusta University, Augusta, GA, USA
| | - Hanping Wu
- Department of Radiology and Imaging, Medical College of Georgia at Augusta University, Augusta, GA, USA
| | - Michael A Winkler
- Department of Radiology and Imaging, Medical College of Georgia at Augusta University, Augusta, GA, USA
| | - Eric J Belin de Chantemèle
- Division of Cardiology, Department of Medicine, Medical College of Georgia at Augusta University, Augusta, GA, USA; Vascular Biology Center, Medical College of Georgia at Augusta University, Augusta, GA, USA
| | - Richard Lee
- Department of Surgery, Medical College of Georgia at Augusta University, Augusta, GA, USA
| | - Ha Won Kim
- Division of Cardiology, Department of Medicine, Medical College of Georgia at Augusta University, Augusta, GA, USA; Vascular Biology Center, Medical College of Georgia at Augusta University, Augusta, GA, USA
| | - Neal L Weintraub
- Division of Cardiology, Department of Medicine, Medical College of Georgia at Augusta University, Augusta, GA, USA; Vascular Biology Center, Medical College of Georgia at Augusta University, Augusta, GA, USA.
| |
Collapse
|
30
|
Berg AR, Hong CG, Svirydava M, Li H, Parel PM, Florida E, O'Hagan R, Pantoja CJ, Lateef SS, Anzenberg P, Harrington CL, Ward G, Zhou W, Sorokin AV, Chen MY, Teague HL, Buckler AJ, Playford MP, Gelfand JM, Mehta NN. Association of S100A8/A9 with lipid-rich necrotic core and treatment with biologic therapy in patients with psoriasis: results from an observational cohort study. J Invest Dermatol 2022; 142:2909-2919. [PMID: 35750149 DOI: 10.1016/j.jid.2022.05.1085] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Revised: 04/21/2022] [Accepted: 05/11/2022] [Indexed: 12/20/2022]
Abstract
Psoriasis is a systemic inflammatory disease with increased risk of atherosclerotic events and premature cardiovascular disease. S100A7, A8/A9, and A12 are protein complexes that are produced by activated neutrophils, monocytes, and keratinocytes in psoriasis. Lipid-rich necrotic core (LRNC) is a high-risk coronary plaque feature previously found to be associated with cardiovascular risk factors and psoriasis severity. LRNC can decrease with biologic therapy, but how this occurs remains unknown. We investigated the relationship between S100 proteins, LRNC, and biologic therapy in psoriasis. S100A8/A9 associated with LRNC in fully adjusted models (β = 0.27, P = 0.009, n=125 psoriasis patients with available coronary CT angiography scans, LRNC analyses, and serum S100A7, S100A8, S100A9, S100A12, and S100A8/A9 levels). At one year, in patients receiving biologic therapy (36 of 73 patients had 1-year CCTA scans available), a 79% reduction in S100A8/A9 levels (-172 (-291.7-26.4) vs -29.9 (-137.9- 50.5) P = 0.04) and a 0.6 mm2 reduction in average LRNC area (0.04 (-0.48-0.77) vs -0.56 (-1.8- 0.13); P = 0.02) were noted. These results highlight the potential role of S100A8/A9 in the development of high-risk coronary plaque in psoriasis.
Collapse
Affiliation(s)
- Alexander R Berg
- Section of Inflammation and Cardiometabolic Diseases, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD
| | - Christin G Hong
- Section of Inflammation and Cardiometabolic Diseases, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD
| | - Maryia Svirydava
- Section of Inflammation and Cardiometabolic Diseases, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD
| | - Haiou Li
- Section of Inflammation and Cardiometabolic Diseases, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD
| | - Philip M Parel
- Section of Inflammation and Cardiometabolic Diseases, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD
| | - Elizabeth Florida
- Section of Inflammation and Cardiometabolic Diseases, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD
| | - Ross O'Hagan
- Section of Inflammation and Cardiometabolic Diseases, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD
| | - Carla J Pantoja
- Section of Inflammation and Cardiometabolic Diseases, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD
| | - Sundus S Lateef
- Section of Inflammation and Cardiometabolic Diseases, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD
| | - Paula Anzenberg
- Section of Inflammation and Cardiometabolic Diseases, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD
| | - Charlotte L Harrington
- Section of Inflammation and Cardiometabolic Diseases, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD
| | - Grace Ward
- St. Jude's Research Hospital, Memphis, TN
| | - Wunan Zhou
- Section of Inflammation and Cardiometabolic Diseases, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD
| | - Alexander V Sorokin
- Section of Inflammation and Cardiometabolic Diseases, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD
| | - Marcus Y Chen
- Section of Inflammation and Cardiometabolic Diseases, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD
| | - Heather L Teague
- Section of Inflammation and Cardiometabolic Diseases, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD
| | | | - Martin P Playford
- Section of Inflammation and Cardiometabolic Diseases, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD
| | - Joel M Gelfand
- Department of Dermatology, University of Pennsylvania, Philadelphia, PA
| | - Nehal N Mehta
- Section of Inflammation and Cardiometabolic Diseases, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD.
| |
Collapse
|
31
|
Schett G, Rahman P, Ritchlin C, McInnes IB, Elewaut D, Scher JU. Psoriatic arthritis from a mechanistic perspective. Nat Rev Rheumatol 2022; 18:311-325. [PMID: 35513599 DOI: 10.1038/s41584-022-00776-6] [Citation(s) in RCA: 73] [Impact Index Per Article: 24.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/29/2022] [Indexed: 12/13/2022]
Abstract
Psoriatic arthritis (PsA) is part of a group of closely related clinical phenotypes ('psoriatic disease') that is defined by shared molecular pathogenesis resulting in excessive, prolonged inflammation in the various tissues affected, such as the skin, the entheses or the joints. Psoriatic disease comprises a set of specific drivers that promote an aberrant immune response and the consequent development of chronic disease that necessitates therapeutic intervention. These drivers include genetic, biomechanical, metabolic and microbial factors that facilitate a robust and continuous mobilization, trafficking and homing of immune cells into the target tissues. The role of genetic variants involved in the immune response, the contribution of mechanical factors triggering an exaggerated inflammatory response (mechanoinflammation), the impact of adipose tissue and altered lipid metabolism and the influence of intestinal dysbiosis in the disease process are discussed. Furthermore, the role of key cytokines, such as IL-23, IL-17 and TNF, in orchestrating the various phases of the inflammatory disease process and as therapeutic targets in PsA is reviewed. Finally, the nature and the mechanisms of inflammatory tissue responses inherent to PsA are summarized.
Collapse
Affiliation(s)
- Georg Schett
- Department of Medicine 3, Friedrich Alexander University Erlangen-Nuremberg and Universitaets-klinikum Erlangen, Erlangen, Germany. .,Deutsches Zentrum Immuntherapie DZI, Friedrich Alexander University Erlangen-Nuremberg and Universitaets-klinikum Erlangen, Erlangen, Germany.
| | - Proton Rahman
- St. Clare's Mercy Hospital, St. John's, Newfoundland, Canada
| | - Christopher Ritchlin
- Division of Allergy, Immunology and Rheumatology, University of Rochester Medical Center Rochester, Rochester, NY, USA
| | - Iain B McInnes
- College of Medical Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
| | - Dirk Elewaut
- VIB-UGent Center for Inflammation Research and Ghent University Hospital, Ghent, Belgium
| | - Jose U Scher
- Department of Medicine, Division of Rheumatology, NYU Grossman School of Medicine, New York, NY, USA
| |
Collapse
|
32
|
Cong L, Xie X, Liu S, Xiang L, Fu X. Genistein promotes M1 macrophage apoptosis and reduces inflammatory response by disrupting miR-21/TIPE2 pathway. Saudi Pharm J 2022; 30:934-945. [PMID: 35903524 PMCID: PMC9315303 DOI: 10.1016/j.jsps.2022.05.009] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2022] [Accepted: 05/20/2022] [Indexed: 11/29/2022] Open
Abstract
Cardiovascular diseases are a major cause of mortality, and vascular injury, a common pathological basis of cardiovascular disease, is deeply correlated with macrophage apoptosis and inflammatory response. Genistein, a type of phytoestrogen, exerts cardiovascular protective activities, but the underlying mechanism has not been fully elucidated. In this study, RAW264.7 cells were treated with genistein, lipopolysaccharide (LPS), nuclear factor-kappa B (NF-κB) inhibitor, and/or protein kinase B (AKT) agonist to determine the role of genistein in apoptosis and inflammation in LPS-stimulated cells. Simultaneously, high fat diet-fed C57BL/6 mice were administered genistein to evaluate the function of genistein on LPS-induced cardiovascular injury mouse model. Here, we demonstrated that LPS obviously increased apoptosis resistance and inflammatory response of macrophages by promoting miR-21 expression, and miR-21 downregulated tumor necrosis factor-α-induced protein 8-like 2 (TIPE2) expression by targeting the coding region. Genistein reduced miR-21 expression by inhibiting NF-κB, then blocked toll-like receptor 4 (TLR4) pathway and AKT phosphorylation dependent on TIPE2, resulting in inhibition of LPS. Our research suggests that miR-21/TIPE2 pathway is involved in M1 macrophage apoptosis and inflammatory response, and genistein inhibits the progression of LPS-induced cardiovascular injury at the epigenetic level via regulating the promoter region of Vmp1 by NF-κB.
Collapse
Affiliation(s)
- Li Cong
- Key Laboratory of Study and Discovery of Small Targeted Molecules of Hunan Province, Changsha 410013, China
- School of Medicine, Hunan Normal University, Changsha 410013, China
- Corresponding authors at: School of Medicine, Hunan Normal University, Changsha 410013, China.
| | - Xiaolin Xie
- School of Medicine, Hunan Normal University, Changsha 410013, China
| | - Sujuan Liu
- School of Medicine, Hunan Normal University, Changsha 410013, China
| | - Liping Xiang
- School of Medicine, Hunan Normal University, Changsha 410013, China
| | - Xiaohua Fu
- School of Medicine, Hunan Normal University, Changsha 410013, China
- Corresponding authors at: School of Medicine, Hunan Normal University, Changsha 410013, China.
| |
Collapse
|
33
|
Scott C, Lateef SS, Hong CG, Dey AK, Manyak GA, Patel NH, Zhou W, Sorokin AV, Abdelrahman K, Uceda D, Teklu M, Wu C, Parel PM, Sandfort V, Chen MY, Mallek M, Ahlman M, Bluemke D, Mehta NN. Inflammation, coronary plaque progression, and statin use: A secondary analysis of the Risk Stratification with Image Guidance of HMG CoA Reductase Inhibitor Therapy (RIGHT) study. Clin Cardiol 2022; 45:622-628. [PMID: 35366378 PMCID: PMC9175258 DOI: 10.1002/clc.23808] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Revised: 02/14/2022] [Accepted: 02/23/2022] [Indexed: 12/02/2022] Open
Abstract
Background Statin treatment is a potent lipid‐lowering therapy associated with decreased cardiovascular risk and mortality. Recent studies including the PARADIGM trial have demonstrated the impact of statins on promoting calcified coronary plaque. Hypothesis The degree of systemic inflammation impacts the amount of increase in coronary plaque calcification over 2 years of statin treatment. Methods A subgroup of 142 participants was analyzed from the Risk Stratification with Image Guidance of HMG CoA Reductase Inhibitor Therapy (RIGHT) study (NCT01212900), who were on statin treatment and underwent cardiac computed tomography angiography (CCTA) at baseline and 2‐year follow‐up. This cohort was stratified by baseline median levels of high‐sensitivity hs‐CRP and analyzed with linear regressions using Stata‐17 (StataCorp). Results In the high versus low hs‐CRP group, patients with higher baseline median hs‐CRP had increased BMI (median [IQR]; 29 [27–31] vs. 27 [24–28]; p < .001), hypertension (59% vs. 41%; p = .03), and LDL‐C levels (97 [77–113] vs. 87 [75–97] mg/dl; p = .01). After 2 years of statin treatment, the high hs‐CRP group had significant increase in dense‐calcified coronary burden versus the low hs‐CRP group (1.27 vs. 0.32 mm2 [100×]; p = .02), beyond adjustment (β = .2; p = .03). Conclusions Statin treatment over 2 years associated with a significant increase in coronary calcification in patients with higher systemic inflammation, as measured by hs‐CRP. These findings suggest that systemic inflammation plays a role in coronary calcification and further studies should be performed to better elucidate these findings.
Collapse
Affiliation(s)
- Colin Scott
- Section of Inflammation and Cardiometabolic Diseases, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Sundus S Lateef
- Section of Inflammation and Cardiometabolic Diseases, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Christin G Hong
- Section of Inflammation and Cardiometabolic Diseases, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Amit K Dey
- Section of Inflammation and Cardiometabolic Diseases, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Grigory A Manyak
- Section of Inflammation and Cardiometabolic Diseases, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Nidhi H Patel
- Section of Inflammation and Cardiometabolic Diseases, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Wunan Zhou
- Section of Inflammation and Cardiometabolic Diseases, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Alexander V Sorokin
- Section of Inflammation and Cardiometabolic Diseases, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Khaled Abdelrahman
- Section of Inflammation and Cardiometabolic Diseases, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Domingo Uceda
- Section of Inflammation and Cardiometabolic Diseases, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Meron Teklu
- Section of Inflammation and Cardiometabolic Diseases, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Colin Wu
- Section of Inflammation and Cardiometabolic Diseases, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Philip M Parel
- Section of Inflammation and Cardiometabolic Diseases, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Veit Sandfort
- Section of Inflammation and Cardiometabolic Diseases, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Marcus Y Chen
- Section of Inflammation and Cardiometabolic Diseases, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Marissa Mallek
- Section of Inflammation and Cardiometabolic Diseases, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Mark Ahlman
- Section of Inflammation and Cardiometabolic Diseases, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland, USA
| | | | - Nehal N Mehta
- Section of Inflammation and Cardiometabolic Diseases, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland, USA
| |
Collapse
|
34
|
Merola JF, McInnes IB, Deodhar AA, Dey AK, Adamstein NH, Quebe-Fehling E, Aassi M, Peine M, Mehta NN. Effect of Secukinumab on Traditional Cardiovascular Risk Factors and Inflammatory Biomarkers: Post Hoc Analyses of Pooled Data Across Three Indications. Rheumatol Ther 2022; 9:935-955. [PMID: 35305260 PMCID: PMC9127026 DOI: 10.1007/s40744-022-00434-z] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Accepted: 02/14/2022] [Indexed: 11/29/2022] Open
Abstract
Background Psoriasis, psoriatic arthritis (PsA), and axial spondyloarthritis (axSpA) are chronic immune-mediated inflammatory diseases (IMIDs) associated with cardiovascular (CV) disease. High-sensitivity C-reactive protein (hsCRP) and, more recently, the neutrophil–lymphocyte ratio (NLR) are important inflammatory biomarkers predictive of CV disease and CV disease-associated mortality. Here, we report the effect of interleukin (IL)-17A inhibition with secukinumab on CV risk parameters in patients with psoriasis, PsA, and axSpA over 1 year of treatment. Methods This was a post hoc analysis of pooled data from phase 3/4 secukinumab studies in psoriasis, PsA, and axSpA. CV-related exclusion criteria included uncontrolled hypertension and congestive heart failure. Traditional risk factors assessed were body mass index (BMI) > 25, high fasting glucose and blood pressure (systolic and diastolic), and high cholesterol (low-density lipoproteins [LDL], total cholesterol/HDL ratio, and triglycerides). Inflammatory CV risk parameters assessed were hsCRP and NLR. Statistical analysis was descriptive. Subgroup analyses were performed in high-risk patients defined as having baseline hsCRP > 4 mg/L (patients with psoriasis) and > 10 mg/L (patients with PsA/axSpA). Results In total, 9197 patients from 19 clinical trials (8 in psoriasis, n = 4742; 5 in PsA, n = 2475; 6 in axSpA, n = 1980) were included. All traditional CV risk parameters remained stable in secukinumab-treated patients through 1 year. Secukinumab rapidly reduced both hsCRP and the NLR compared with placebo at week 12 (psoriasis) or week 16 (PsA/axSpA) in the overall population and in high-risk patients (all P < 0.01). This reduction was maintained for at least 1 year of secukinumab therapy in all indications. Conclusions Secukinumab led to a rapid and sustained reduction in hsCRP and the NLR in patients with IMIDs with a high systemic inflammatory burden. Traditional CV risk factors remained stable for at least 1 year in patients with psoriasis, PsA, and axSpA. Taken together, secukinumab had a favorable effect on systemic inflammation without impact on traditional CV risk factors. Trials Registration ClinicalTrials.gov, NCT01365455, NCT01358578, NCT01406938, NCT01555125, NCT01636687, NCT02752776, NCT02074982, NCT02826603, NCT01752634, NCT01989468, NCT02294227, NCT02404350, NCT02745080, NCT01863732, NCT01649375, NCT02008916, NCT02159053, NCT02896127, NCT02696031. Supplementary Information The online version contains supplementary material available at 10.1007/s40744-022-00434-z.
Collapse
Affiliation(s)
- Joseph F Merola
- Brigham and Women's Hospital, Harvard Medical School, Boston, USA
| | | | | | - Amit K Dey
- Section of Inflammation and Cardiometabolic Diseases, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | | | | | | | | | - Nehal N Mehta
- Section of Inflammation and Cardiometabolic Diseases, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, 20892, USA.
| |
Collapse
|
35
|
Silveira Rossi JL, Barbalho SM, Reverete de Araujo R, Bechara MD, Sloan KP, Sloan LA. Metabolic syndrome and cardiovascular diseases: Going beyond traditional risk factors. Diabetes Metab Res Rev 2022; 38:e3502. [PMID: 34614543 DOI: 10.1002/dmrr.3502] [Citation(s) in RCA: 243] [Impact Index Per Article: 81.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Accepted: 08/28/2021] [Indexed: 12/14/2022]
Abstract
Metabolic syndrome (MS) is a chronic non-infective syndrome characterised clinically by a set of vascular risk factors that include insulin resistance, hypertension, abdominal obesity, impaired glucose metabolism, and dyslipidaemia. These risk factors are due to a pro-inflammatory state, oxidative stress, haemodynamic dysfunction, and ischaemia, which overlap in 'dysmetabolic' patients. This review aimed to evaluate the relationship between the traditional components of MS with cardiovascular disease (CVD), inflammation, and oxidative stress. MEDLINE-PubMed, EMBASE, and Cochrane databases were searched. Chronic low-grade inflammatory states and metaflammation are often accompanied by metabolic changes directly related to CVD incidence, such as diabetes mellitus, hypertension, and obesity. Moreover, the metaflammation is characterised by an increase in the serum concentration of pro-inflammatory cytokines, mainly interleukin-1 β (IL-1β), IL-6, and tumour necrosis factor-α (TNF-α), originating from the chronically inflamed adipose tissue and associated with oxidative stress. The increase of reactive oxygen species overloads the antioxidant systems causing post-translational alterations of proteins, lipids, and DNA leading to oxidative stress. Hyperglycaemia contributes to the increase in oxidative stress and the production of advanced glycosylation end products (AGEs) which are related to cellular and molecular dysfunction. Oxidative stress and inflammation are associated with cellular senescence and CVD. CVD should not be seen only as being triggered by classical MS risk factors. Atherosclerosis is a multifactorial pathological process with several triggering and aetiopathogenic mechanisms. Its medium and long-term repercussions, however, invariably constitute a significant cause of morbidity and mortality. Implementing preventive and therapeutic measures against oxy-reductive imbalances and metaflammation states has unquestionable potential for favourable clinical outcomes in cardiovascular medicine.
Collapse
Affiliation(s)
- João Leonardo Silveira Rossi
- Department of Biochemistry and Pharmacology, School of Medicine, University of Marília, Marília, São Paulo, Brazil
| | - Sandra Maria Barbalho
- Department of Biochemistry and Pharmacology, School of Medicine, University of Marília, Marília, São Paulo, Brazil
- Postgraduate Program in Structural and Functional Interactions in Rehabilitation - University of Marília, Marília, São Paulo, Brazil
- School of Food and Technology of Marilia, Marilia, São Paulo, Brazil
| | - Renan Reverete de Araujo
- Department of Biochemistry and Pharmacology, School of Medicine, University of Marília, Marília, São Paulo, Brazil
| | - Marcelo Dib Bechara
- Department of Biochemistry and Pharmacology, School of Medicine, University of Marília, Marília, São Paulo, Brazil
| | | | - Lance Alan Sloan
- Texas Institute for Kidney and Endocrine Disorders, Lufkin, Texas, USA
- University of Texas Medical Branch, Galveston, Texas, USA
| |
Collapse
|
36
|
Li Y, Yu Z, Liu Y, Wang T, Liu Y, Bai Z, Ren Y, Ma H, Bao T, Lu H, Wang R, Yang L, Yan N, Yan R, Jia S, Zhang X, Wang H. Dietary α-Linolenic Acid-Rich Flaxseed Oil Ameliorates High-Fat Diet-Induced Atherosclerosis via Gut Microbiota-Inflammation-Artery Axis in ApoE−/− Mice. Front Cardiovasc Med 2022; 9:830781. [PMID: 35295260 PMCID: PMC8918482 DOI: 10.3389/fcvm.2022.830781] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Accepted: 01/24/2022] [Indexed: 12/12/2022] Open
Abstract
Atherosclerosis (AS) is closely associated with abnormally chronic low-grade inflammation and gut dysbiosis. Flaxseed oil (FO) rich in omega-3 polyunsaturated fatty acids (PUFAs), which are mainly composed of alpha-linolenic acid (ALA, 18:3 omega-3), has been demonstrated to exhibit pleiotropic benefits in chronic metabolic diseases. However, the impact of dietary ALA-rich FO on AS and its associated underlying mechanisms remain poorly understood. Thus, the present study was designed as two phases to investigate the effects in atherosclerotic Apolipoprotein E (ApoE)−/− mice. In the initial portion, the ApoE−/− mice were randomly allocated to three groups: control group (CON), model group (MOD), and FO-fed model group (MOD/FO) and were treated for 12 weeks. The second phase used antibiotic (AB)-treated ApoE−/− mice were divided into two groups: AB-treated model group (AB/MOD) and FO-fed AB-treated model group (AB/FO). In the results, the dietary ALA-rich FO administration ameliorated atherosclerotic lesion, as well as the parameters of AS (body weights (BWs) and the total bile acids (TBA). Chronic systemic/vascular inflammatory cytokines and in situ macrophages (Mψs) were reduced with FO intervention. In addition, the FO improved the gut integrity and permeability by decreasing the plasma lipopolysaccharide (LPS). Moreover, gut dysbiosis and metabolites [short-chain fatty acids (SCFAs) and bile acids (BAs)] in AS were modulated after FO treatment. Intriguingly, during an AB-treated condition, a significantly weakened amelioration of FO-treated on AS proposed that the intestinal microbiota contributed to the FO effects. A correlation analysis showed close relationships among gut bacteria, metabolites, and inflammation. Collectively, these results suggested that the dietary ALA-rich FO ameliorated the AS in ApoE−/− mice via the gut microbiota-inflammation-artery axis.
Collapse
Affiliation(s)
- Yiwei Li
- School of Basic Medical Sciences, Ningxia Medical University, Yinchuan, China
| | - Zhi Yu
- Department of Anesthesiology, People's Hospital of Ningxia Hui Autonomous Region, Yinchuan, China
| | - Yuanyuan Liu
- School of Basic Medical Sciences, Ningxia Medical University, Yinchuan, China
| | - Ting Wang
- School of Basic Medical Sciences, Ningxia Medical University, Yinchuan, China
| | - Yajuan Liu
- Clinical Medical College, Ningxia Medical University, Yinchuan, China
- Department of Cardiovascular Diseases, Heart Centre, General Hospital of Ningxia Medical University, Yinchuan, China
- Ningxia Key Laboratory of Vascular Injury and Repair Research, Ningxia Medical University, Yinchuan, China
| | - Zhixia Bai
- Clinical Medical College, Ningxia Medical University, Yinchuan, China
- Department of Cardiovascular Diseases, Heart Centre, General Hospital of Ningxia Medical University, Yinchuan, China
- Ningxia Key Laboratory of Vascular Injury and Repair Research, Ningxia Medical University, Yinchuan, China
| | - Yi Ren
- Clinical Medical College, Ningxia Medical University, Yinchuan, China
| | - Huiyan Ma
- Clinical Medical College, Ningxia Medical University, Yinchuan, China
- Department of Cardiovascular Diseases, Heart Centre, General Hospital of Ningxia Medical University, Yinchuan, China
- Ningxia Key Laboratory of Vascular Injury and Repair Research, Ningxia Medical University, Yinchuan, China
| | - Ting Bao
- Clinical Medical College, Ningxia Medical University, Yinchuan, China
| | - Haixia Lu
- Department of Anesthesiology, People's Hospital of Ningxia Hui Autonomous Region, Yinchuan, China
| | - Rui Wang
- Clinical Medical College, Ningxia Medical University, Yinchuan, China
| | - Libo Yang
- Department of Cardiovascular Diseases, Heart Centre, General Hospital of Ningxia Medical University, Yinchuan, China
- Ningxia Key Laboratory of Vascular Injury and Repair Research, Ningxia Medical University, Yinchuan, China
| | - Ning Yan
- Department of Cardiovascular Diseases, Heart Centre, General Hospital of Ningxia Medical University, Yinchuan, China
- Ningxia Key Laboratory of Vascular Injury and Repair Research, Ningxia Medical University, Yinchuan, China
| | - Ru Yan
- Department of Cardiovascular Diseases, Heart Centre, General Hospital of Ningxia Medical University, Yinchuan, China
- Ningxia Key Laboratory of Vascular Injury and Repair Research, Ningxia Medical University, Yinchuan, China
| | - Shaobin Jia
- Department of Cardiovascular Diseases, Heart Centre, General Hospital of Ningxia Medical University, Yinchuan, China
- Ningxia Key Laboratory of Vascular Injury and Repair Research, Ningxia Medical University, Yinchuan, China
| | - Xiaoxia Zhang
- College of Traditional Chinese Medicine, Ningxia Medical University, Yinchuan, China
- *Correspondence: Xiaoxia Zhang
| | - Hao Wang
- School of Basic Medical Sciences, Ningxia Medical University, Yinchuan, China
- Hao Wang
| |
Collapse
|
37
|
Gonzalez-Cantero A, Teklu M, Sorokin AV, Prussick R, González-Cantero J, Martin-Rodriguez JL, Patel N, Parel PM, Manyak GA, Teague HL, Rodante JA, Keel A, Pérez-Hortet C, Sanchéz-Moya AI, Jiménez N, Ballester A, Solis J, Fernandez-Friera L, Barderas MG, Gonzalez-Calvin JL, Jaen P, Playford MP, Dey AK, Gelfand JM, Mehta NN. Subclinical Liver Disease Is Associated with Subclinical Atherosclerosis in Psoriasis: Results from Two Observational Studies. J Invest Dermatol 2022; 142:88-96. [PMID: 34293354 DOI: 10.1016/j.jid.2021.05.034] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Revised: 05/10/2021] [Accepted: 05/17/2021] [Indexed: 12/13/2022]
Abstract
Psoriasis is associated with a higher risk of liver diseases. We investigated the impact of hepatic steatosis (European cohort) and hepatic inflammation (United States cohort) on subclinical atherosclerosis. In the European cohort (n = 76 psoriasis participants and 76 controls), nonalcoholic fatty liver disease, assessed by the sonographic hepatorenal index, was more prevalent in psoriasis than in controls (61% vs. 45%; P = 0.04). Participants with psoriasis with nonalcoholic fatty liver disease had a higher prevalence of subclinical atherosclerosis (ultrasonographic presence of plaque in femoral or carotid arteries) than participants with psoriasis without nonalcoholic fatty liver disease (61% vs. 23%; P = 0.006) and controls with nonalcoholic fatty liver disease (61% vs. 32%; P < 0.05). Sonographic hepatorenal index was a determinant of subclinical atherosclerosis in psoriasis (OR = 3.5; P = 0.01). In the United States cohort (n = 162 participants with psoriasis who underwent positron emission tomography and coronary computed tomography angiography), those with high hepatic 2-[fluorine-18]fluoro-2-deoxy-D-glucose uptake had higher noncalcified (1.3 [0.49 mm2] vs. 1.0 [0.40 mm2]), fibrofatty (0.23 [0.15 mm2] vs. 0.11 [0.087 mm2]), and lipid-rich necrotic core (4.3 [2.3 mm2] vs. 3.0 [1.7 mm2]) coronary burden (all P < 0.001). Hepatic 2-[fluorine-18]fluoro-2-deoxy-D-glucose uptake associated with noncalcified (β = 0.28; P < 0.001), fibrofatty (β = 0.49; P < 0.001), and lipid-rich necrotic core (β = 0.28; P = 0.003) burden. These results show the downstream cardiovascular effects of subclinical liver disease in psoriasis.
Collapse
Affiliation(s)
- Alvaro Gonzalez-Cantero
- Department of Dermatology, Hospital Universitario Ramon y Cajal, Madrid, Spain; Facultad de Medicina, Universidad Francisco de Vitoria, Madrid, Spain
| | - Meron Teklu
- National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Alexander V Sorokin
- National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Ronald Prussick
- The Department of Dermatology, The George Washington School of Medicine & Health Sciences, Washington, District of Columbia, USA
| | | | | | - Nidhi Patel
- National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Philip M Parel
- National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Grigory A Manyak
- National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Heather L Teague
- National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Justin A Rodante
- National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Andrew Keel
- National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland, USA
| | | | - Ana I Sanchéz-Moya
- Department of Dermatology, Complejo Hospitalario de Toledo, Toledo, Spain
| | - Natalia Jiménez
- Department of Dermatology, Hospital Universitario Ramon y Cajal, Madrid, Spain
| | - Asunción Ballester
- Department of Dermatology, Hospital Universitario Ramon y Cajal, Madrid, Spain
| | - Jorge Solis
- Department of Cardiology, Hospital Universitario 12 de Octubre, Madrid, Spain
| | | | - María G Barderas
- Department of Vascular Physiopathology, Hospital Nacional de Parapléjicos, SESCAM, Toledo, Spain
| | | | - Pedro Jaen
- Department of Dermatology, Hospital Universitario Ramon y Cajal, Madrid, Spain
| | - Martin P Playford
- National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Amit K Dey
- National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Joel M Gelfand
- Department of Biostatistics, Epidemiology & Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA; Department of Dermatology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Nehal N Mehta
- National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland, USA.
| |
Collapse
|
38
|
Wang E, Cesano A, Butterfield LH, Marincola F. Improving the therapeutic index in adoptive cell therapy: key factors that impact efficacy. J Immunother Cancer 2021; 8:jitc-2020-001619. [PMID: 33023983 PMCID: PMC7539608 DOI: 10.1136/jitc-2020-001619] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/16/2020] [Indexed: 12/13/2022] Open
Abstract
The therapeutic index (TI) is a quantitative assessment of a drug safety proportional to its effectiveness. The estimation is intuitive when the engagement of the product with its target is dependent on stable chemistry and predictable pharmacokinetics as is the case for small molecules or antibodies. But for therapeutics with complex biodistribution and context-dependent potency such as adoptive cell therapy (ACT) products, TI estimations need to consider a broader array of factors. These include product-dependent variability such as functional fitness, unpredictable pharmacokinetics due to non-specific trapping, sequestration and extravasation into normal tissues and variable rates of in vivo expansion. In the case of solid malignancies, additional modifiers dependent on individual tumor immune biology may affect pharmacodynamics, including differential trafficking to benign compared with cancer tissue, hampered engagement with target cells, immune suppression and cellular dysfunction due to unfavorable metabolic conditions. Here, we propose a patient-specific assessment of factors affecting on-tumor from off-tumor activity in disparate immunologic environments that impact ACT’s clinical efficacy and may favorably balance the TI. for ACT products.
Collapse
Affiliation(s)
- Ena Wang
- Allogene Therapeutics, San Francisco, California, USA
| | | | - Lisa H Butterfield
- Research, Parker Institute for Cancer Immunotherapy, San Francisco, California, USA.,Microbiology and Immunology, University of California San Francisco, San Francisco, California, USA
| | | |
Collapse
|
39
|
McInnes IB, Gravallese EM. Immune-mediated inflammatory disease therapeutics: past, present and future. Nat Rev Immunol 2021; 21:680-686. [PMID: 34518662 PMCID: PMC8436867 DOI: 10.1038/s41577-021-00603-1] [Citation(s) in RCA: 154] [Impact Index Per Article: 38.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/29/2021] [Indexed: 02/07/2023]
Abstract
Immune-mediated inflammatory diseases are common and clinically diverse. Although they are currently incurable, the therapeutic armamentarium for immune-mediated inflammatory diseases has been transformed in the past two decades. We have moved from the wide application of broad-spectrum immune modulators to the routine use of agents with exquisite specificity, arising from monoclonal and molecular biotechnology and more recently from highly targeted medicinal chemistry. Here we describe key advances and lessons that drove this remarkable progress and thereafter reflect on the next steps in this ongoing journey.
Collapse
Affiliation(s)
- Iain B McInnes
- College of Medical Veterinary and Life Sciences, University of Glasgow, Glasgow, UK.
| | - Ellen M Gravallese
- Division of Rheumatology, Inflammation, and Immunity, Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| |
Collapse
|
40
|
Nast A, Altenburg A, Augustin M, Boehncke WH, Härle P, Klaus J, Koza J, Mrowietz U, Ockenfels HM, Philipp S, Reich K, Rosenbach T, Schlaeger M, Schmid-Ott G, Sebastian M, von Kiedrowski R, Weberschock T, Dressler C. German S3-Guideline on the treatment of Psoriasis vulgaris, adapted from EuroGuiDerm - Part 2: Treatment monitoring and specific clinical or comorbid situations. J Dtsch Dermatol Ges 2021; 19:1092-1115. [PMID: 34288477 DOI: 10.1111/ddg.14507] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Accepted: 03/06/2021] [Indexed: 12/20/2022]
Affiliation(s)
- Alexander Nast
- Department of Dermatology, Venereology and Allergology, Division of Evidence-Based Medicine (dEBM), Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Andreas Altenburg
- Dermatology, Venereology and Allergology, Immunology Center, Dessau Municipal Hospital, Dessau, Germany
| | - Matthias Augustin
- German Center for Health Services Research in Dermatology (CVderm), Institute for Health Services Research in Dermatology and Nursing (IVDP), University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany
| | - Wolf-Henning Boehncke
- Department of Dermatology and Venereology, University Hospital of Geneva, Geneva, Switzerland
| | | | | | | | - Ulrich Mrowietz
- Psoriasis Center, Department of Dermatology, Venereology, Allergology, University Hospital Schleswig-Holstein, Campus Kiel, Kiel, Germany
| | | | | | - Kristian Reich
- Center for Translational Research in Inflammatory Skin Diseases, Institute for Health Services Research in Dermatology and Nursing, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | | | | | | | | | | | - Tobias Weberschock
- Department of Dermatology, Venereology and Allergology, University Hospital Frankfurt, Frankfurt/Main, Germany and Working group Evidence-based Medicine Frankfurt, Institute for General Practice, Goethe University Frankfurt, Frankfurt/Main, Germany
| | - Corinna Dressler
- Department of Dermatology, Venereology and Allergology, Division of Evidence-Based Medicine (dEBM), Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| |
Collapse
|
41
|
Nast A, Altenburg A, Augustin M, Boehncke WH, Härle P, Klaus J, Koza J, Mrowietz U, Ockenfels HM, Philipp S, Reich K, Rosenbach T, Schlaeger M, Schmid-Ott G, Sebastian M, von Kiedrowski R, Weberschock T, Dressler C. Deutsche S3-Leitlinie zur Therapie der Psoriasis vulgaris, adaptiert von EuroGuiDerm - Teil 2: Therapiemonitoring, besondere klinische Situationen und Komorbidität. J Dtsch Dermatol Ges 2021; 19:1092-1117. [PMID: 34288473 DOI: 10.1111/ddg.14507_g] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Accepted: 03/06/2021] [Indexed: 12/12/2022]
Affiliation(s)
- Alexander Nast
- Klinik für Dermatologie, Venerologie und Allergologie, Division of Evidence-Based Medicine (dEBM), Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin
| | - Andreas Altenburg
- Dermatologie, Venerologie und Allergologie, Immunologisches Zentrum, Städtisches Klinikum Dessau
| | - Matthias Augustin
- Kompetenzzentrum Versorgungsforschung in der Dermatologie (CVderm), Institut für Versorgungsforschung in der Dermatologie und bei Pflegeberufen (IVDP), Universitätsklinikum Eppendorf, Hamburg
| | | | | | | | | | - Ulrich Mrowietz
- Psoriasis-Zentrum, Klinik für Dermatologie, Venerologie, Allergologie, Universitätsklinikum Schleswig-Holstein, Campus Kiel
| | | | | | - Kristian Reich
- Zentrum für Translationale Forschung bei entzündlichen Hauterkrankungen, Institut für Versorgungsforschung in der Dermatologie und bei Pflegeberufen, Universitätsmedizin Hamburg-Eppendorf
| | | | | | | | | | | | - Tobias Weberschock
- Klinik für Dermatologie, Venerologie und Allergologie, Universitätsklinikum Frankfurt, Frankfurt/Main und Arbeitsgruppe EbM Frankfurt, Institut für Allgemeinmedizin, Goethe-Universität Frankfurt, Frankfurt
| | - Corinna Dressler
- Klinik für Dermatologie, Venerologie und Allergologie, Division of Evidence-Based Medicine (dEBM), Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin
| |
Collapse
|
42
|
Teklu M, Parel PM, Mehta NN. Psoriasis and Cardiometabolic Diseases: The Impact of Inflammation on Vascular Health. PSORIASIS-TARGETS AND THERAPY 2021; 11:99-108. [PMID: 34322373 PMCID: PMC8312325 DOI: 10.2147/ptt.s320016] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/13/2021] [Accepted: 07/06/2021] [Indexed: 12/24/2022]
Abstract
Psoriasis is a common chronic inflammatory condition associated with a higher risk of cardiovascular disease. Psoriasis confers a dose-dependent increase in risk for the metabolic syndrome and its components. The metabolic syndrome and its components have been associated with higher coronary atherosclerosis in psoriasis and cardiovascular events in the general population. In this review, we discuss the role of inflammation and psoriasis in cardiometabolic diseases with a focus on the metabolic syndrome and its components. We highlight the relationship between psoriasis and important cardiovascular risk factors encompassed by obesity, dyslipidemia, insulin resistance and hypertension. Furthermore, we briefly highlight literature on anti-inflammatory therapies and their impact on the components of the metabolic syndrome as well as directly quantified coronary atherosclerosis burden.
Collapse
Affiliation(s)
- Meron Teklu
- National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Philip M Parel
- National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Nehal N Mehta
- National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
43
|
Visser MJE, Tarr G, Pretorius E. Thrombosis in Psoriasis: Cutaneous Cytokine Production as a Potential Driving Force of Haemostatic Dysregulation and Subsequent Cardiovascular Risk. Front Immunol 2021; 12:688861. [PMID: 34335591 PMCID: PMC8324086 DOI: 10.3389/fimmu.2021.688861] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Accepted: 07/06/2021] [Indexed: 12/18/2022] Open
Abstract
Psoriasis (PsO) is a common T cell-mediated inflammatory disorder of the skin with an estimated prevalence of 2%. The condition manifests most commonly as erythematous plaques covered with scales. The aetiology of PsO is multifactorial and disease initiation involves interactions between environmental factors, susceptibility genes, and innate and adaptive immune responses. The underlying pathology is mainly driven by interleukin-17. In addition, various inflammatory mediators from specific T helper (TH) cell subsets, namely TH1, TH17, and TH22, are overexpressed in cutaneous lesions and may also be detected in the peripheral blood of psoriatic patients. Moreover, these individuals are also at greater risk, compared to the general population, of developing multiple comorbid conditions. Cardiovascular disease (CVD) has been recognised as a prominent comorbidity of PsO. A potential mechanism contributing to this association may be the presence of a hypercoagulable state in these individuals. Inflammation and coagulation are closely related. The presence of chronic, low-grade systemic inflammation may promote thrombosis – one of the major determinants of CVD. A pro-inflammatory milieu may induce the expression of tissue factor, augment platelet activity, and perturb the vascular endothelium. Altogether, these changes will result in a prothrombotic state. In this review, we describe the aetiology of PsO, as well as the pathophysiology of the condition. We also consider its relationship to CVD. Given the systemic inflammatory nature of PsO, we evaluate the potential contribution of prominent inflammatory mediators (implicated in PsO pathogenesis) to establishing a prothrombotic state in psoriatic patients.
Collapse
Affiliation(s)
- Maria J E Visser
- Department of Physiological Sciences, Faculty of Science, Stellenbosch University, Stellenbosch, South Africa
| | - Gareth Tarr
- Department of Physiological Sciences, Faculty of Science, Stellenbosch University, Stellenbosch, South Africa.,Division of Rheumatology, Institute of Orthopaedics and Rheumatology, Winelands Mediclinic Orthopaedic Hospital, Stellenbosch, South Africa
| | - Etheresia Pretorius
- Department of Physiological Sciences, Faculty of Science, Stellenbosch University, Stellenbosch, South Africa
| |
Collapse
|
44
|
Park C, Lee H, Kwon CY, Kim GY, Jeong JW, Kim SO, Choi SH, Jeong SJ, Noh JS, Choi YH. Loganin Inhibits Lipopolysaccharide-Induced Inflammation and Oxidative Response through the Activation of the Nrf2/HO-1 Signaling Pathway in RAW264.7 Macrophages. Biol Pharm Bull 2021; 44:875-883. [PMID: 34078820 DOI: 10.1248/bpb.b21-00176] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Inflammation caused by the excessive secretion of inflammatory mediators in abnormally activated macrophages promotes many diseases along with oxidative stress. Loganin, a major iridoid glycoside isolated from Cornus officinalis, has recently been reported to exhibit anti-inflammatory and antioxidant effects, whereas the underlying mechanism has not yet been fully clarified. Therefore, the aim of the present study is to investigate the effect of loganin on inflammation and oxidative stress in lipopolysaccharide (LPS)-stimulated RAW264.7 macrophages. Our results indicated that loganin treatment markedly attenuated the LPS-mediated phagocytic activity and release of nitric oxide (NO) and prostaglandin E2, which was associated with decreased the expression of inducible NO synthase and cyclooxygenase-2. In addition, loganin suppressed the expression and their extracellular secretion of LPS-induced pro-inflammatory cytokines, such as tumor necrosis factor-α and interleukin-1β. Furthermore, loganin abolished reactive oxygen species (ROS) generation, and promoted the activation of nuclear factor-E2-related factor 2 (Nrf2) and the expression of heme oxygenase-1 (HO-1) in LPS-stimulated macrophages. However, zinc protoporphyrin, a selective HO-1 inhibitor, reversed the loganin-mediated suppression of pro-inflammatory cytokines in LPS-treated macrophages. In conclusion, our findings suggest that the upregulation of the Nrf2/HO-1 signaling pathway is concerned at least in the protective effect of loganin against LPS-mediated inflammatory and oxidative stress, and that loganin can be a potential functional agent to prevent inflammatory and oxidative damage.
Collapse
Affiliation(s)
- Cheol Park
- Division of Basic Sciences, College of Liberal Studies, Dong-eui University
| | - Hyesook Lee
- Anti-Aging Research Center, Dong-eui University.,Department of Biochemistry, Dong-eui University College of Korean Medicine
| | - Chan-Young Kwon
- Department of Oriental Neuropsychiatry, Dong-eui University College of Korean Medicine
| | - Gi-Young Kim
- Laboratory of Immunobiology, Department of Marine Life Sciences, Jeju National University
| | - Jin-Woo Jeong
- Nakdonggang National Institute of Biological Resources
| | - Sung Ok Kim
- Department of Food Science & Biotechnology, College of Engineering, Kyungsung University
| | | | - Soon-Jeong Jeong
- Department of Dental Hygiene, College of Health Science, Youngsan University
| | - Jeong Sook Noh
- Department of Food Science & Nutrition, Tongmyong University
| | - Yung Hyun Choi
- Anti-Aging Research Center, Dong-eui University.,Department of Biochemistry, Dong-eui University College of Korean Medicine
| |
Collapse
|
45
|
Kwon DH, Kim GY, Cha HJ, Kim S, Kim HS, Hwang HJ, Choi YH. Nargenicin A1 attenuates lipopolysaccharide-induced inflammatory and oxidative response by blocking the NF-κB signaling pathway. EXCLI JOURNAL 2021; 20:968-982. [PMID: 34267609 PMCID: PMC8278209 DOI: 10.17179/excli2021-3506] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/12/2021] [Accepted: 05/26/2021] [Indexed: 12/04/2022]
Abstract
Inflammation caused by the excessive production of pro-inflammatory mediators and cytokines in abnormally activated macrophages promotes the initiation and progression of many diseases along with oxidative stress. Previous studies have suggested that nargenicin A1, an antibacterial macrolide isolated from Nocardia sp. may be a potential treatment for inflammatory responses and oxidative stress, but the detailed mechanisms are still not well studied. In this study, we investigated the inhibitory effect of nargenicin A1 on inflammatory and oxidative stress in lipopolysaccharide (LPS)-stimulated RAW 264.7 macrophages and zebrafish (Danio rerio) models. Our results indicated that nargenicin A1 treatment significantly inhibited LPS-induced release of pro-inflammatory mediators including nitric oxide (NO) and prostaglandin E2, which was associated with decreased inducible NO synthase and cyclooxygenase-2 expression. In addition, nargenicin A1 attenuated the LPS-induced expression of pro-inflammatory cytokines, such as tumor necrosis factor (TNF)-α, interleukin (IL)-1β, IL-6, and monocyte chemotactic protein-1, reducing their extracellular secretion. Nargenicin A1 also suppressed LPS-induced generation of reactive oxygen species. Moreover, nargenicin A1 abolished the LPS-mediated nuclear translocation of nuclear factor-kappa B (NF-κB) and the degradation of inhibitor IκB-α, indicating that nargenicin A1 exhibited anti-inflammatory effects by inhibiting the NF-κB signaling pathway. Furthermore, nargenicin A1 showed strong protective effects against NO and ROS production in LPS-injected zebrafish larvae. In conclusion, our findings suggest that nargenicin A1 ameliorates LPS-induced anti-inflammatory and antioxidant effects by downregulating the NF-κB signaling pathway, and that nargenicin A1 can be a potential functional agent to prevent inflammatory- and oxidative-mediated damage.
Collapse
Affiliation(s)
- Da Hye Kwon
- Anti‐Aging Research Center, Dong‐eui University, Busan, Republic of Korea
- Department of Biochemistry, Dong‐eui University College of Korean Medicine, Busan, Republic of Korea
| | - Gi-Young Kim
- Department of Marine Life Science, School of Marine Biomedical Sciences, Jeju National University, Jeju, Republic of Korea
| | - Hee-Jae Cha
- Department of Parasitology and Genetics, College of Medicine, Kosin University, Busan, Republic of Korea
| | - Suhkmann Kim
- Department of Chemistry, College of Natural Sciences, Pusan National University, Busan, Republic of Korea
| | - Heui-Soo Kim
- Department of Biological Sciences, College of Natural Sciences, Pusan National University, Busan, Republic of Korea
| | - Hye-Jin Hwang
- Department of Food and Nutrition, College of Nursing, Healthcare Sciences & Human Ecology, Dong-eui University, Busan, Republic of Korea
| | - Yung Hyun Choi
- Anti‐Aging Research Center, Dong‐eui University, Busan, Republic of Korea
- Department of Biochemistry, Dong‐eui University College of Korean Medicine, Busan, Republic of Korea
| |
Collapse
|
46
|
Park C, Cha HJ, Lee H, Kim GY, Choi YH. The regulation of the TLR4/NF-κB and Nrf2/HO-1 signaling pathways is involved in the inhibition of lipopolysaccharide-induced inflammation and oxidative reactions by morroniside in RAW 264.7 macrophages. Arch Biochem Biophys 2021; 706:108926. [PMID: 34029560 DOI: 10.1016/j.abb.2021.108926] [Citation(s) in RCA: 57] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Revised: 05/16/2021] [Accepted: 05/17/2021] [Indexed: 02/07/2023]
Abstract
Morroniside, a major iridoid glycoside isolated from Cornus officinalis, has a variety of beneficial pharmacological properties. Although morroniside has recently been reported to exhibit anti-inflammatory and antioxidant effects, the detailed mechanism has not yet been fully elucidated. In this study, we investigated the inhibitory effect of morroniside on inflammatory and oxidative stress activated by lipopolysaccharide (LPS) in RAW 264.7 macrophages. Our results indicated that morroniside pretreatment significantly inhibited the LPS-induced phagocytic activity and release of pro-inflammatory factors, which was associated with blocking the expression of their regulatory genes. Morroniside also markedly suppressed the expression of myeloid differentiation factor 88 as well as Toll-like receptor 4 (TLR4), and attenuated the translocation of nuclear factor-κB (NF-κB) to the nucleus in LPS-treated RAW 264.7 macrophages. Furthermore, morroniside prevented the binding of LPS to the TLR4 on the cell surface. In addition, morroniside abolished reactive oxygen species (ROS) generation, and enhanced the expression of heme oxygenase-1 (HO-1) following activation of nuclear factor-E2-related factor 2 (Nrf2) in LPS-stimulated RAW 264.7 macrophages. However, zinc protoporphyrin, a specific inhibitor of HO-1, reversed the morroniside-mediated inhibition of inflammatory response in LPS-treated RAW 264.7 macrophages. In conclusion, our findings suggest that morroniside exerts LPS-induced anti-inflammatory and antioxidant effects by targeting the TLR4/NF-κB and Nrf2/HO-1 signaling pathways in RAW 264.7 macrophages. Taken together, our findings suggest that morroniside interacted structurally and electrochemically with TLR4/MD2 complex, consequently can be a potential functional agent to prevent inflammatory and oxidative damage.
Collapse
Affiliation(s)
- Cheol Park
- Division of Basic Sciences, College of Liberal Studies, Dong-eui University, Busan, 47340, Republic of Korea
| | - Hee-Jae Cha
- Department of Parasitology and Genetics, College of Medicine, Kosin University, Busan, 49104, Republic of Korea
| | - Hyesook Lee
- Anti-Aging Research Center, Dong-eui University, Busan, 47340, Republic of Korea; Department of Biochemistry, Dong-eui University College of Korean Medicine, Busan, 47227, Republic of Korea
| | - Gi-Young Kim
- Department of Marine Life Sciences, School of Marine Biomedical Sciences, Jeju National University, Jeju, 63243, Republic of Korea
| | - Yung Hyun Choi
- Anti-Aging Research Center, Dong-eui University, Busan, 47340, Republic of Korea; Department of Biochemistry, Dong-eui University College of Korean Medicine, Busan, 47227, Republic of Korea.
| |
Collapse
|
47
|
Ji SY, Cha HJ, Molagoda IMN, Kim MY, Kim SY, Hwangbo H, Lee H, Kim GY, Kim DH, Hyun JW, Kim HS, Kim S, Jin CY, Choi YH. Suppression of Lipopolysaccharide-Induced Inflammatory and Oxidative Response by 5-Aminolevulinic Acid in RAW 264.7 Macrophages and Zebrafish Larvae. Biomol Ther (Seoul) 2021; 29:685-696. [PMID: 33820881 PMCID: PMC8551728 DOI: 10.4062/biomolther.2021.030] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2021] [Revised: 03/11/2021] [Accepted: 03/15/2021] [Indexed: 11/05/2022] Open
Abstract
In this study, we investigated the inhibitory effect of 5-aminolevulinic acid (ALA), a heme precursor, on inflammatory and oxidative stress activated by lipopolysaccharide (LPS) in RAW 264.7 macrophages by estimating nitric oxide (NO), prostaglandin E2 (PGE2), cytokines, and reactive oxygen species (ROS). We also evaluated the molecular mechanisms through analysis of the expression of their regulatory genes, and further evaluated the anti-inflammatory and antioxidant efficacy of ALA against LPS in the zebrafish model. Our results indicated that ALA treatment significantly attenuated the LPS-induced release of pro-inflammatory mediators including NO and PGE2, which was associated with decreased inducible NO synthase and cyclooxygenase-2 expression. ALA also inhibited the LPS-induced expression of pro-inflammatory cytokines, such as tumor necrosis factor (TNF)-α, interleukin (IL)-1β, and IL-6, reducing their extracellular secretion. Additionally, ALA abolished ROS generation, improved the mitochondrial mass, and enhanced the expression of heme oxygenase-1 (HO-1) and the activation of nuclear translocation of nuclear factor-E2-related factor 2 (Nrf2) in LPS-stimulated RAW 264.7 macrophages. However, zinc protoporphyrin, a specific inhibitor of HO-1, reversed the ALA-mediated inhibition of pro-inflammatory cytokines production and activation of mitochondrial function in LPS-treated RAW 264.7 macrophages. Furthermore, ALA significantly abolished the expression of LPS-induced pro-inflammatory mediators and cytokines, and showed strong protective effects against NO and ROS production in zebrafish larvae. In conclusion, our findings suggest that ALA exerts LPS-induced anti-inflammatory and antioxidant effects by upregulating the Nrf2/HO-1 signaling pathway, and that ALA can be a potential functional agent to prevent inflammatory and oxidative damage.
Collapse
Affiliation(s)
- Seon Yeong Ji
- Anti-Aging Research Center, Dong-eui University, Busan 47340, Republic of Korea.,Department of Biochemistry, College of Korean Medicine, Dong-eui University, Busan 47227, Republic of Korea
| | - Hee-Jae Cha
- Department of Parasitology and Genetics, College of Medicine, Kosin University, Busan 49104, Republic of Korea
| | | | - Min Yeong Kim
- Anti-Aging Research Center, Dong-eui University, Busan 47340, Republic of Korea.,Department of Biochemistry, College of Korean Medicine, Dong-eui University, Busan 47227, Republic of Korea
| | - So Young Kim
- Anti-Aging Research Center, Dong-eui University, Busan 47340, Republic of Korea.,Department of Biochemistry, College of Korean Medicine, Dong-eui University, Busan 47227, Republic of Korea
| | - Hyun Hwangbo
- Anti-Aging Research Center, Dong-eui University, Busan 47340, Republic of Korea.,Department of Biochemistry, College of Korean Medicine, Dong-eui University, Busan 47227, Republic of Korea
| | - Hyesook Lee
- Anti-Aging Research Center, Dong-eui University, Busan 47340, Republic of Korea.,Department of Biochemistry, College of Korean Medicine, Dong-eui University, Busan 47227, Republic of Korea
| | - Gi-Young Kim
- Department of Marine Life Sciences, Jeju National University, Jeju 63243, Republic of Korea
| | - Do-Hyung Kim
- Department of Aquatic Life Medicine, Pukyong National University, Busan 48513, Republic of Korea
| | - Jin Won Hyun
- Department of Biochemistry, College of Medicine, Jeju National University, Jeju 63243, Republic of Korea
| | - Heui-Soo Kim
- Department of Biological Sciences, Pusan National University, Busan 46241, Republic of Korea
| | - Suhkmann Kim
- Department of Chemistry, Pusan National University, Busan 46241, Republic of Korea
| | - Cheng-Yun Jin
- School of Pharmaceutical Sciences, Zhengzhou University, Henan 450001, China
| | - Yung Hyun Choi
- Anti-Aging Research Center, Dong-eui University, Busan 47340, Republic of Korea.,Department of Biochemistry, College of Korean Medicine, Dong-eui University, Busan 47227, Republic of Korea
| |
Collapse
|
48
|
Rachfal AW, Grant SFA, Schwartz SS. The Diabetes Syndrome - A Collection of Conditions with Common, Interrelated Pathophysiologic Mechanisms. Int J Gen Med 2021; 14:923-936. [PMID: 33776471 PMCID: PMC7987256 DOI: 10.2147/ijgm.s305156] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Accepted: 03/08/2021] [Indexed: 11/23/2022] Open
Abstract
The four basic pathophysiologic mechanisms which damage the β-cell within diabetes (ie, genetic and epigenetic changes, inflammation, an abnormal environment, and insulin resistance [IR]) also contribute to cell and tissue damage and elevate the risk of developing all typical diabetes-related complications. Genetic susceptibility to damage from abnormal external and internal environmental factors has been described including inflammation and IR. All these mechanisms can promote epigenetic changes, and in total, these pathophysiologic mechanisms interact and react with each other to cause damage to cells and tissues ultimately leading to disease. Importantly, these pathophysiologic mechanisms also serve to link other common conditions including cancer, dementia, psoriasis, atherosclerotic cardiovascular disease (ASCVD), nonalcoholic fatty liver disease (NAFLD), and nonalcoholic steatohepatitis (NASH). The "Diabetes Syndrome", an overarching group of interrelated conditions linked by these overlapping mechanisms, can be viewed as a conceptual framework that can facilitate understanding of the inter-relationships of superficially disparate conditions. Recognizing the association of the conditions within the Diabetes Syndrome due to common pathophysiologies has the potential to provide both benefit to the patient (eg, prevention, early detection, precision medicine) and to the advancement of medicine (eg, driving education, research, and dynamic decision-based medical practice).
Collapse
Affiliation(s)
| | - Struan F A Grant
- Center for Spatial and Functional Genomics, Division of Human Genetics, Children’s Hospital of Philadelphia, Philadelphia, PA, USA
- Department of Pediatrics, University of Pennsylvania, Perlman School of Medicine, Philadelphia, PA, USA
- Department of Genetics, University of Pennsylvania, Perlman School of Medicine, Philadelphia, PA, USA
| | - Stanley S Schwartz
- Stanley Schwartz MD, LLC, Main Line Health System, Wynnewood, PA, USA
- University of Pennsylvania, Perlman School of Medicine, Philadelphia, PA, USA
| |
Collapse
|
49
|
Edigin E, Kaul S, Eseaton PO, Ojemolon PE, Patel A, Manadan A. Psoriasis does not worsen outcomes in patients admitted for ischemic stroke: an analysis of the National Inpatient Sample. J Investig Med 2021; 69:994-998. [PMID: 33723000 DOI: 10.1136/jim-2020-001678] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/25/2021] [Indexed: 11/03/2022]
Abstract
Psoriasis is a chronic inflammatory state associated with an increased risk of cardiometabolic diseases, stroke, and mortality. Although psoriasis increases the risk of ischemic stroke, whether outcomes, including mortality, are adversely affected is unknown.This study aims to compare inpatient mortality of patients admitted for ischemic stroke with and without psoriasis. The secondary outcome measures were hospital length of stay (LOS), total hospital charges, odds of receiving tissue plasminogen activator (TPA), and mechanical thrombectomy between both groups.Data were obtained from the National Inpatient Sample (NIS) 2016 and 2017 databases using the International Classification of Diseases, Tenth Revision, Clinical Modification codes. Multivariable logistic and linear regression analysis were used accordingly to account for confounders of the outcomes.The combined 2016 and 2017 NIS database comprised over 71 million discharges. Of these, ischemic stroke accounted for 525,570 hospitalizations and 2425 (0.5%) had a concomitant diagnosis of psoriasis. Patients hospitalized for ischemic stroke with coexisting psoriasis did not have a difference in inpatient mortality (3.5% vs 5.5%; p=0.285) compared with those without psoriasis. However, psoriasis cohort had shorter LOS (5.0 vs 5.7 days; p=0.029) and lower total hospital charges ($60,471 vs $70,246; p=0.003) compared with the non-psoriasis cohort. The odds of receiving TPA and undergoing mechanical thrombectomy were not different in both groups.Inpatient mortality, odds of receiving TPA, and undergoing mechanical thrombectomy in patients who had an ischemic stroke with or without psoriasis were not different. However, patients with psoriasis had a significantly shorter LOS and lower hospital charges.
Collapse
Affiliation(s)
- Ehizogie Edigin
- Department of Internal Medicine, John H Stroger Hospital of Cook County, Chicago, Illinois, USA
| | - Subuhi Kaul
- Department of Internal Medicine, John H Stroger Hospital of Cook County, Chicago, Illinois, USA
| | | | | | - Axi Patel
- Department of Internal Medicine, John H Stroger Hospital of Cook County, Chicago, Illinois, USA
| | - Augustine Manadan
- Rheumatology, John H Stroger Hospital of Cook County, Chicago, Illinois, USA.,Rheumatology, Rush University Medical Center, Chicago, Illinois, USA
| |
Collapse
|
50
|
Teklu M, Zhou W, Kapoor P, Patel N, Dey AK, Sorokin AV, Manyak GA, Teague HL, Erb-Alvarez JA, Sajja A, Abdelrahman KM, Reddy AS, Uceda DE, Lateef SS, Shanbhag SM, Scott C, Prakash N, Svirydava M, Parel P, Rodante JA, Keel A, Siegel EL, Chen MY, Bluemke DA, Playford MP, Gelfand JM, Mehta NN. Metabolic syndrome and its factors are associated with noncalcified coronary burden in psoriasis: An observational cohort study. J Am Acad Dermatol 2021; 84:1329-1338. [PMID: 33383084 DOI: 10.1016/j.jaad.2020.12.044] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Revised: 12/10/2020] [Accepted: 12/16/2020] [Indexed: 12/18/2022]
Abstract
BACKGROUND Psoriasis is associated with a heightened risk of cardiovascular disease and higher prevalence of metabolic syndrome. OBJECTIVE Investigate the effect of metabolic syndrome and its factors on early coronary artery disease assessed as noncalcified coronary burden by coronary computed tomography angiography in psoriasis. METHODS This cross-sectional study consisted of 260 participants with psoriasis and coronary computed tomography angiography characterization. Metabolic syndrome was defined according to the harmonized International Diabetes Federation criteria. RESULTS Of the 260 participants, 80 had metabolic syndrome (31%). The metabolic syndrome group had a higher burden of cardiometabolic disease, systemic inflammation, noncalcified coronary burden, and high-risk coronary plaque. After adjusting for Framingham risk score, lipid-lowering therapy, and biologic use, metabolic syndrome (β = .31; P < .001) and its individual factors of waist circumference (β = .33; P < .001), triglyceride levels (β = .17; P = .005), blood pressure (β = .18; P = .005), and fasting glucose (β = .17; P = .009) were significantly associated with noncalcified coronary burden. After adjusting for all other metabolic syndrome factors, blood pressure and waist circumference remained significantly associated with noncalcified coronary burden. LIMITATIONS Observational nature with limited ability to control for confounders. CONCLUSIONS In psoriasis, individuals with metabolic syndrome had more cardiovascular disease risk factors, systemic inflammation, and noncalcified coronary burden. Efforts to increase metabolic syndrome awareness in psoriasis should be undertaken to reduce the heightened cardiovascular disease risk.
Collapse
Affiliation(s)
- Meron Teklu
- National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland
| | - Wunan Zhou
- National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland
| | - Promita Kapoor
- National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland
| | - Nidhi Patel
- National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland
| | - Amit K Dey
- National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland
| | - Alexander V Sorokin
- National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland
| | - Grigory A Manyak
- National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland
| | - Heather L Teague
- National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland
| | - Julie A Erb-Alvarez
- National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland
| | - Aparna Sajja
- Department of Internal Medicine, Johns Hopkins University Medical Center, Baltimore, Maryland
| | - Khaled M Abdelrahman
- National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland
| | - Aarthi S Reddy
- National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland
| | - Domingo E Uceda
- National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland
| | - Sundus S Lateef
- National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland
| | - Sujata M Shanbhag
- National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland
| | - Colin Scott
- National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland
| | - Nina Prakash
- National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland
| | - Maryia Svirydava
- National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland
| | - Philip Parel
- National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland
| | - Justin A Rodante
- National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland
| | - Andrew Keel
- National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland
| | - Evan L Siegel
- Department of Rheumatology, Arthritis and Rheumatism Associates, Rockville, Maryland
| | - Marcus Y Chen
- National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland
| | - David A Bluemke
- Department of Radiology, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin
| | - Martin P Playford
- National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland
| | - Joel M Gelfand
- Department of Dermatology, Perelman School of Medicine, Philadelphia, Pennsylvania; Department of Biostatistics, Epidemiology and Informatics, Perelman School of Medicine, Philadelphia, Pennsylvania
| | - Nehal N Mehta
- National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland.
| |
Collapse
|