1
|
Su X, Yu H, Lei Q, Chen X, Tong Y, Zhang Z, Yang W, Guo Y, Lin L. Systemic lupus erythematosus: pathogenesis and targeted therapy. MOLECULAR BIOMEDICINE 2024; 5:54. [PMID: 39472388 PMCID: PMC11522254 DOI: 10.1186/s43556-024-00217-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2024] [Accepted: 10/16/2024] [Indexed: 11/02/2024] Open
Abstract
Systemic lupus erythematosus (SLE) is a multifaceted autoimmune disorder characterized by dysregulated immune responses and autoantibody production, which affects multiple organs and varies in clinical presentation and disease severity. The development of SLE is intricate, encompassing dysregulation within the immune system, a collapse of immunological tolerance, genetic susceptibilities to the disease, and a variety of environmental factors that can act as triggers. This review provides a comprehensive discussion of the pathogenesis and treatment strategies of SLE and focuses on the progress and status of traditional and emerging treatment strategies for SLE. Traditional treatment strategies for SLE have mainly employed non-specific approaches, including cytotoxic and immunosuppressive drugs, antimalarials, glucocorticoids, and NSAIDs. These strategies are effective in mitigating the effects of the disease, but they are not a complete cure and are often accompanied by adverse reactions. Emerging targeted therapeutic drugs, on the other hand, aim to control and treat SLE by targeting B and T cells, inhibiting their activation and function, as well as the abnormal activation of the immune system. A deeper understanding of the pathogenesis of SLE and the exploration of new targeted treatment strategies are essential to advance the treatment of this complex autoimmune disease.
Collapse
Affiliation(s)
- Xu Su
- Medical Research Center, College of Medicine, The Third People's Hospital of Chengdu (Affiliated Hospital of Southwest Jiaotong University, Southwest Jiaotong University, Chengdu, 610031, Sichuan, China
| | - Hui Yu
- Department of Urology, The Third People's Hospital of Chengdu, The Affiliated Hospital of Southwest Jiaotong University, Chengdu, 610014, China
| | - Qingqiang Lei
- Center of Bone Metabolism and Repair, Department of Wound Repair and Rehabilitation Medicine, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Daping Hospital, Army Medical University, Chongqing, 400000, China
| | - Xuerui Chen
- Medical Research Center, College of Medicine, The Third People's Hospital of Chengdu (Affiliated Hospital of Southwest Jiaotong University, Southwest Jiaotong University, Chengdu, 610031, Sichuan, China
| | - Yanli Tong
- Université Paris Cité, INSERM U1151, CNRS UMR8253, Institut Necker Enfants Malades, Paris, F-75015, France
| | - Zhongyang Zhang
- Department of Health Technology, The Danish National Research Foundation and Villum Foundation's Center IDUN, Technical University of Denmark, Kgs. Lyngby, Denmark
| | - Wenyong Yang
- Medical Research Center, College of Medicine, The Third People's Hospital of Chengdu (Affiliated Hospital of Southwest Jiaotong University, Southwest Jiaotong University, Chengdu, 610031, Sichuan, China.
- Department of Neurosurgery, The Third People's Hospital of Chengdu, The Affiliated Hospital of Southwest Jiaotong University, Chengdu, 610014, China.
| | - Yuanbiao Guo
- Medical Research Center, College of Medicine, The Third People's Hospital of Chengdu (Affiliated Hospital of Southwest Jiaotong University, Southwest Jiaotong University, Chengdu, 610031, Sichuan, China.
| | - Liangbin Lin
- Medical Research Center, College of Medicine, The Third People's Hospital of Chengdu (Affiliated Hospital of Southwest Jiaotong University, Southwest Jiaotong University, Chengdu, 610031, Sichuan, China.
- Obesity and Metabolism Medicine-Engineering Integration Laboratory, Department of General Surgery, The Third People's Hospital of Chengdu, The Affiliated Hospital of Southwest Jiaotong University, Chengdu, 610031, China.
- The Center of Gastrointestinal and Minimally Invasive Surgery, Department of General Surgery, The Third People's Hospital of Chengdu, The Affiliated Hospital of Southwest Jiaotong University, Chengdu, 610031, China.
| |
Collapse
|
2
|
Leerach N, Ngaosuwan K, Mahikul W. Regional variations in serum IL-35 levels and association with systemic lupus erythematosus: a systematic review and meta-analysis. Sci Rep 2024; 14:24820. [PMID: 39438756 PMCID: PMC11496805 DOI: 10.1038/s41598-024-76375-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Accepted: 10/14/2024] [Indexed: 10/25/2024] Open
Abstract
Interleukin (IL)-35 is an anti-inflammatory cytokine that regulates autoimmune diseases, including systemic lupus erythematosus (SLE). However, the association between the cytokine and disease may vary by geographical region. This study performed a meta-analysis to quantitatively assess the correlation between the serum IL-35 levels in SLE patients and sub-group analyses were conducted. Four main electronic databases-Scopus, Embase, Science Direct, PubMed-were searched for relevant studies. After a database search, Endnote software was used to find and remove duplicate studies. Random-effects models were used to estimate standard mean differences in serum/plasma IL-35 levels by Hedges' g with 95% confidence intervals (CIs). Publication bias was assessed with funnel plots, and risk of bias was assessed according to the Newcastle-Ottawa Scale (NOS). Sixteen studies met the eligibility criteria and were included in a qualitative review; data from 15 studies were included in the meta-analysis. Total IL-35 levels (pg/mL) did not differ among patients with active SLE and healthy controls (Hedges's g: 0.22, 95% CI - 0.51, 0.95, p = 0.55). Sub-group analysis revealed that IL-35 levels in patients with active SLE were lower than in healthy controls in Chinese studies (Hedges's g: - 3.11, 95% CI - 5.72, - 0.51), but not in non-Chinese studies (Hedges's g: 1.63, 95% CI - 0.31, 3.57). This regional difference was statistically significant (p < 0.01). The analysis comparing patients with inactive SLE and healthy controls showed a similar trend. This study suggests that serum IL-35 levels are lower in patients with SLE in studies from China, but not other regions. However, standardized protocols with large sample sizes are needed to confirm these findings.
Collapse
Affiliation(s)
- Nontaphat Leerach
- Princess Srisavangavadhana College of Medicine, Chulabhorn Royal Academy, Bangkok, 10210, Thailand.
| | - Kanchana Ngaosuwan
- Princess Srisavangavadhana College of Medicine, Chulabhorn Royal Academy, Bangkok, 10210, Thailand
| | - Wiriya Mahikul
- Princess Srisavangavadhana College of Medicine, Chulabhorn Royal Academy, Bangkok, 10210, Thailand.
| |
Collapse
|
3
|
Ilavarasan R, Arunadevi R, Kusuma G, Gaidhani SN, Thenmozhi M, Manikandan N. Evaluation of Anti-arthritic and in-vitro Anti-inflammatory activity of Vaisvanara Churna. JOURNAL OF ETHNOPHARMACOLOGY 2024; 337:118774. [PMID: 39218128 DOI: 10.1016/j.jep.2024.118774] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Revised: 08/20/2024] [Accepted: 08/30/2024] [Indexed: 09/04/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Vaisvanara Churna used traditionally for the treatment of Amavata (Rheumatoid arthritis), Shotaprasamana (Anti-inflammatory) and as Saraka (Laxative). AIM Aim of the study is to evaluate anti arthritic activity and in vitro anti-inflammatory potential of Vaisvanara Churna in experimental animals. MATERIALS AND METHODS In-vitro anti-inflammatory activity of aqueous extract of Vaisvanara Churna (100-500 μg/ml) was evaluated by using membrane stabilization methods. Anti arthritic activity was evaluated by using 0.1 ml of Complete Freund's Adjuvant (CFA) injected into sub plantar surface of left hind paw of each wistar rat on day 1 followed by treatment with Vaisvanara Churna at various dose levels (450, 900, and 1800 mg/kg b.w) and standard drug Prednisolone (5 mg/kg) for 21 days. The following parameters namely change in the body weight of animals, paw volume, ankle joint thickness were measured at 0, 3, 8, 17 & 21 day intervals and radiographic changes were assessed. In addition to this, response to painful stimuli by application of forced pressure was measured by using Pressure Application Measurement (PAM) method. RESULTS In-vitro anti-inflammatory activity Vaisvanara Churna exhibited dose dependent membrane stabilizing activity. Treatment with Vaisvanara Churna showed significant (p < 0.05) inhibition of paw edema, reduction in ankle joint thickness and increase in the body weight of wistar albino rats was observed. There is a significant increase (p < 0.001) in the latency of limb withdrawal response, reduction in the organ indices (spleen and thymus) were noted. CONCLUSION This study demonstrates that Vaisvanara Churna possesses in vitro anti inflammatory and anti-arthritic potential and supports its folklore use in the treatment of arthritis.
Collapse
Affiliation(s)
- R Ilavarasan
- Department of Pharmacology, Captain Srinivasa Murthy Central Ayurveda Research Institute, Central Council for Research in Ayurvedic Sciences, Ministry of AYUSH, Govt. of India, Chennai, 600 106, India
| | - R Arunadevi
- Department of Pharmacology, Captain Srinivasa Murthy Central Ayurveda Research Institute, Central Council for Research in Ayurvedic Sciences, Ministry of AYUSH, Govt. of India, Chennai, 600 106, India.
| | - G Kusuma
- Department of Pharmacology, Captain Srinivasa Murthy Central Ayurveda Research Institute, Central Council for Research in Ayurvedic Sciences, Ministry of AYUSH, Govt. of India, Chennai, 600 106, India
| | - Sudesh N Gaidhani
- Central Council for Research in Ayurvedic Sciences, Ministry of AYUSH, New Delhi, Govt. of India, 110058, India
| | - M Thenmozhi
- Department of Pharmacology, Captain Srinivasa Murthy Central Ayurveda Research Institute, Central Council for Research in Ayurvedic Sciences, Ministry of AYUSH, Govt. of India, Chennai, 600 106, India
| | - N Manikandan
- Department of Pharmacology, Captain Srinivasa Murthy Central Ayurveda Research Institute, Central Council for Research in Ayurvedic Sciences, Ministry of AYUSH, Govt. of India, Chennai, 600 106, India
| |
Collapse
|
4
|
Lee S, Son SK, Cho E, Yoo S, Jang EA, Kwak SH. Protective Role of Astaxanthin in Regulating Lipopolysaccharide-Induced Inflammation and Apoptosis in Human Neutrophils. Curr Issues Mol Biol 2024; 46:8567-8575. [PMID: 39194721 DOI: 10.3390/cimb46080504] [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: 06/17/2024] [Revised: 07/22/2024] [Accepted: 07/30/2024] [Indexed: 08/29/2024] Open
Abstract
Astaxanthin, a keto-carotenoid, is known to have potent antioxidant properties. This study aims to investigate the anti-inflammatory effect of astaxanthin and its mechanism in human neutrophils. The effects of astaxanthin on lipopolysaccharide (LPS)-stimulated human neutrophils were investigated in vitro. Neutrophils were isolated from healthy volunteers and stimulated with LPS in the presence and absence of astaxanthin. We assessed cytokine production, signaling pathway activation via mitogen-activated protein kinases (MAPKs) and nuclear factor kappa B (NF-κB), and apoptosis. Astaxanthin's impact was evaluated at different concentrations, and both pretreatment and cotreatment protocols were tested. The results demonstrated that astaxanthin significantly reduced the production of pro-inflammatory cytokines TNF-α and IL-1β in LPS-stimulated neutrophils. It effectively inhibited the phosphorylation of ERK1/2 MAPK, without notably affecting p38 MAPK or NF-κB pathways. Furthermore, astaxanthin promoted apoptosis in neutrophils, counteracting the apoptosis-delaying effects of LPS. These effects were more pronounced with pretreatment. In conclusion, astaxanthin has protective effects on inflammatory responses in neutrophils by reducing cytokine production and enhancing apoptosis while selectively modulating intracellular signaling pathways. Astaxanthin demonstrates significant potential as a therapeutic agent in the management of severe inflammatory conditions.
Collapse
Affiliation(s)
- Seongheon Lee
- Department of Anesthesiology and Pain Medicine, Chonnam National University Hospital, Chonnam National University Medical School, Gwangju 61469, Republic of Korea
| | - Sung Kuk Son
- Department of Anesthesiology and Pain Medicine, Chonnam National University Hospital, Chonnam National University Medical School, Gwangju 61469, Republic of Korea
| | - Eunye Cho
- Department of Anesthesiology and Pain Medicine, Chonnam National University Hospital, Chonnam National University Medical School, Gwangju 61469, Republic of Korea
| | - Sungah Yoo
- Department of Anesthesiology and Pain Medicine, Chonnam National University Hospital, Chonnam National University Medical School, Gwangju 61469, Republic of Korea
| | - Eun-A Jang
- Department of Anesthesiology and Pain Medicine, Chonnam National University Hospital, School of Dentistry, Chonnam National University, Gwangju 61186, Republic of Korea
| | - Sang Hyun Kwak
- Department of Anesthesiology and Pain Medicine, Chonnam National University Hospital, Chonnam National University Medical School, Gwangju 61469, Republic of Korea
| |
Collapse
|
5
|
Kim HB, Go EJ, Baek JS. Effect of hot-melt extruded Morus alba leaves on intestinal microflora and epithelial cells. Heliyon 2024; 10:e23954. [PMID: 38332870 PMCID: PMC10851307 DOI: 10.1016/j.heliyon.2023.e23954] [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: 09/22/2023] [Revised: 11/30/2023] [Accepted: 12/18/2023] [Indexed: 02/10/2024] Open
Abstract
Although rutin and isoquercitrin have many effects, they are insoluble substances, making it difficult to obtain pure substances. This study was to investigate whether Morus alba leaves containing rutin and isoquercitrin could improve intestinal health by making a sustained-release formulation through a hot-melt extrusion (HME) process with improved stability and solubility and determine whether it could upregulate the balance of intestinal microorganisms and intestinal epithelial cells. A sustained-release formulation was prepared by the HME process using Morus alba leaves and a hydrophilic polymer matrix. Antibacterial activities of pathogenic microorganisms (Escherichia coli, Streptococcus aureus, Enterococcus faecalis) and proliferative effect of probiotics (Lactobacillus rhamnosus, Pediococcus pentosaceus) were tested against intestinal microorganisms. Regarding intestinal epithelial cells, a co-culture model of Caco-2 cells and RAW 264.7 cells was used. It was confirmed that the extrudate exhibited high antibacterial activities against pathogenic microorganisms and affected the proliferation of probiotics. Furthermore, after inducing inflammation through LPS, it recovered transepithelial electrical resistance-increased levels of tight junction proteins and decreased expression levels of pro-inflammatory cytokines. HME of Morus alba leaves containing rutin and isoquercitrin can upregulate intestinal microbial balance and intestinal epithelial cells.
Collapse
Affiliation(s)
- Hyun Bok Kim
- National Institute of Agricultural Sciences, RDA, Wanju 55365, South Korea
| | - Eun Ji Go
- Department of Bio-Health Convergence, Kangwon National University, Chuncheon 24341, South Korea
| | - Jong-Suep Baek
- Department of Bio-Health Convergence, Kangwon National University, Chuncheon 24341, South Korea
- Department of Bio-Functional Materials, Kangwon National University, Samcheok 25949. South Korea
- BeNatureBioLab, Chuncheon 24206, South Korea
| |
Collapse
|
6
|
Jha A, Joseph J, Prabhu SB, Chaudhary A, Yadav B, Mathew J. Utility of peripheral blood monocyte subsets, circulating immune complexes and serum cytokines in assessment of SLE activity: an observational, cross-sectional study. Clin Rheumatol 2024; 43:209-217. [PMID: 38040877 DOI: 10.1007/s10067-023-06832-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2023] [Revised: 11/19/2023] [Accepted: 11/23/2023] [Indexed: 12/03/2023]
Abstract
INTRODUCTION SLE disease measurements by current standards are less than perfect. Monocytes and their subsets are part of innate immunity, and one of our objectives was to look at their role in SLE disease activity. We also looked at the common serum cytokines and the role of circulating immune complex (CIC) estimation in the assessment of disease activity. METHODS We conducted a single-centre observational cross-sectional study of SLE patients with active and inactive disease as the comparison arms. Blood samples were collected for (a) peripheral blood monocyte separation and flowcytometric analysis of monocyte subsets based on CD14 and CD16 surface markers, and (b) ELISA for serum cytokines and CIC estimation. Results were analysed in terms of the difference in medians between the active and inactive disease groups using the Mann-Whitney U test (non-normally distributed data). RESULTS The absolute monocyte count was lower in the active group than the inactive group (median (IQR) of 329 (228.5) vs. 628 (257)/microliter, p = 0.001). The frequency (%) of the intermediate monocyte subset showed a trend towards an increase in active disease (median (IQR) of 15.10% (9.65) vs. 11.85% (8.00), p = 0.09). It also had a significant positive correlation to the SLEDAI scores (r = 0.33, p = 0.046). The mean fluorescence intensity (MFI) of CD163, expressed primarily by intermediate subsets, was increased, and CD11c MFI was reduced in active disease. Serum TNF-a level was elevated in active disease (median (IQR) of 38 (48.5) pg/ml vs. 9 (48.5) pg/ml, p = 0.042). CIC ELISA at an optimal cut-off of 10 meq/ml provided an area under the curve (AUC) of 0.85 for detecting active SLE. CONCLUSION Peripheral blood monocytes are depleted in active disease. The intermediate monocyte subset may have a role in disease activity. TNF-alpha correlated modestly with disease activity. CIC estimation by ELISA may be used in addition to or as an alternative to current standards of laboratory tests for the serological assessment of activity.
Collapse
Affiliation(s)
- Avanish Jha
- Department of Clinical Immunology and Rheumatology, Christian Medical College, Vellore, Tamil Nadu, India.
| | - Josna Joseph
- Department of Clinical Immunology and Rheumatology, Christian Medical College, Vellore, Tamil Nadu, India
| | - Savit B Prabhu
- Wellcome trust research laboratory, Christian Medical College, Vellore, Tamil Nadu, India
| | - Anita Chaudhary
- Wellcome trust research laboratory, Christian Medical College, Vellore, Tamil Nadu, India
| | - Bijesh Yadav
- Department of Biostatistics, Christian Medical College, Vellore, Tamil Nadu, India
| | - John Mathew
- Department of Clinical Immunology and Rheumatology, Christian Medical College, Vellore, Tamil Nadu, India
| |
Collapse
|
7
|
Mackieh R, Al-Bakkar N, Kfoury M, Okdeh N, Pietra H, Roufayel R, Legros C, Fajloun Z, Sabatier JM. Unlocking the Benefits of Fasting: A Review of its Impact on Various Biological Systems and Human Health. Curr Med Chem 2024; 31:1781-1803. [PMID: 38018193 DOI: 10.2174/0109298673275492231121062033] [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: 08/02/2023] [Revised: 09/26/2023] [Accepted: 10/17/2023] [Indexed: 11/30/2023]
Abstract
Fasting has gained significant attention in recent years for its potential health benefits in various body systems. This review aims to comprehensively examine the effects of fasting on human health, specifically focusing on its impact on different body's physiological systems. The cardiovascular system plays a vital role in maintaining overall health, and fasting has shown promising effects in improving cardiovascular health markers such as blood pressure, cholesterol levels, and triglyceride levels. Additionally, fasting has been suggested to enhance insulin sensitivity, promote weight loss, and improve metabolic health, thus offering potential benefits to individuals with diabetes and metabolic disorders. Furthermore, fasting can boost immune function, reduce inflammation, enhance autophagy, and support the body's defense against infections, cancer, and autoimmune diseases. Fasting has also demonstrated a positive effect on the brain and nervous system. It has been associated with neuroprotective properties, improving cognitive function, and reducing the risk of neurodegenerative diseases, besides the ability of increasing the lifespan. Hence, understanding the potential advantages of fasting can provide valuable insights for individuals and healthcare professionals alike in promoting health and wellbeing. The data presented here may have significant implications for the development of therapeutic approaches and interventions using fasting as a potential preventive and therapeutic strategy.
Collapse
Affiliation(s)
- Rawan Mackieh
- Department of Biology, Faculty of Sciences, Lebanese University, Campus Michel Slayman Ras Maska, Tripoli 1352, Lebanon
| | - Nadia Al-Bakkar
- Faculty of Health Sciences, College of Life Sciences, Beirut Arab University, Beirut Campus, P.O. Box 11 50 20, Riad El Solh, Beirut 11072809, Lebanon
| | - Milena Kfoury
- Department of Biology, Faculty of Sciences, Lebanese University, Campus Michel Slayman Ras Maska, Tripoli 1352, Lebanon
| | - Nathalie Okdeh
- Department of Biology, Faculty of Sciences, Lebanese University, Campus Michel Slayman Ras Maska, Tripoli 1352, Lebanon
| | - Hervé Pietra
- Association Esprit Jeûne & Fasting Spirit, 226, Chemin du Pélican, Toulon 83000, France
| | - Rabih Roufayel
- College of Engineering and Technology, American University of the Middle East, Hadiya, Kuwait
| | - Christian Legros
- Univ Angers, INSERM, CNRS, MITOVASC, Team 2 CarMe, SFR ICAT, Angers 49000, France
| | - Ziad Fajloun
- Department of Biology, Faculty of Sciences, Lebanese University, Campus Michel Slayman Ras Maska, Tripoli 1352, Lebanon
- Laboratory of Applied Biotechnology (LBA3B), Azm Center for Research in Biotechnology and its Applications, EDST, Lebanese University, Tripoli 1300, Lebanon
| | - Jean-Marc Sabatier
- Aix-- Marseille Université, CNRS, INP, Inst Neurophysiopathol, Marseille 13385, France
| |
Collapse
|
8
|
Alsbihawi TQ, Zare Ebrahimabad M, Seyedhosseini FS, Davoodi H, Abdolahi N, Nazari A, Mohammadi S, Yazdani Y. Altered expression of miR-125a and dysregulated cytokines in systemic lupus erythematosus: Unveiling diagnostic and prognostic markers. World J Exp Med 2023; 13:102-114. [PMID: 38173550 PMCID: PMC10758663 DOI: 10.5493/wjem.v13.i5.102] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Revised: 09/18/2023] [Accepted: 10/23/2023] [Indexed: 12/19/2023] Open
Abstract
BACKGROUND Systemic lupus erythematosus (SLE) is a chronic autoimmune disorder impacting multiple organs, influenced by genetic factors, especially those related to the immune system. However, there is a need for new biomarkers in SLE. MicroRNA-125a (miR-125a) levels are decreased in T cells, B cells, and dendritic cells of SLE patients. MiR-125a plays a regulatory role in controlling the levels of tumor necrosis factor-alpha (TNF-α) and interleukin 12 (IL-12), which are crucial pro-inflammatory cytokines in SLE pathogenesis. AIM To assess the levels of miR-125a, IL-12, and TNF-α in SLE patients' plasma, evaluating their diagnostic and prognostic value. METHODS The study included 100 healthy individuals, 50 newly diagnosed (ND), and 50 SLE patients undergoing treatment. The patients were monitored for a duration of 24 wk to observe and record instances of relapses. MiR-125a expression was measured using real-time reverse transcription polymerase chain reaction, while ELISA kits were used to assess IL-12 and TNF-α production. RESULTS The results showed significantly reduced miR-125a expression in SLE patients compared to healthy individuals, with the lowest levels in ND patients. TNF-α and IL-12 expression levels were significantly elevated in SLE patients, especially in the early stages of the disease. Receiver operating characteristic curve analyses, and Cox-Mantel Log-rank tests indicated miR-125a, TNF-α, and IL-12 as proper diagnostic biomarkers for SLE. A negative correlation was found between plasma miR-125a expression and IL-12/TNF-α levels in SLE patients. CONCLUSION Decreased miR-125a levels may be involved in the development of SLE, while elevated levels of IL-12 and TNF-α contribute to immune dysregulation. These findings offer new diagnostic and prognostic markers for SLE. Moreover, the negative correlation observed suggests an interaction between miR-125a, TNF-α, and IL-12. Further research is necessary to uncover the underlying mechanisms that govern these relationships.
Collapse
Affiliation(s)
- Tagreed Qassim Alsbihawi
- Department of Immunology, Faculty of Medicine, Golestan University of Medical Sciences, Gorgan 49341-74515, Iran
| | - Mojtaba Zare Ebrahimabad
- Metabolic Disorders Research Center, Golestan University of Medical Sciences, Gorgan 49341-74515, Iran
| | | | - Homa Davoodi
- Department of Immunology, Golestan University of Medical Sciences, Gorgan 4934174515, Iran
| | - Nafiseh Abdolahi
- Golestan Rheumatology Research Center, Golestan University of Medical Sciences, Gorgan 49341-74515, Iran
| | - Alireza Nazari
- Department of Surgery, Faculty of Medicine, Rafsanjan University of Medical Sciences, Rafsanjan 7717933777, Iran
| | - Saeed Mohammadi
- Golestan Research Center of Gastroenterology and Hepatology, Golestan University of Medical Sciences, Gorgan 49341-74515, Iran
| | - Yaghoub Yazdani
- Laboratory Sciences Research Center, Golestan University of Medical Sciences, Gorgan 49341-74515, Iran
| |
Collapse
|
9
|
Khakdan F, Javanmard AS, Shahmoradipour P, Jahromi MJ. The fluctuations of expression profiles of critical genes in the miRNA maturation process and pro-and anti-inflammatory cytokines in the pathogenesis and progression of multiple sclerosis. Mol Biol Rep 2023; 50:9405-9416. [PMID: 37823932 DOI: 10.1007/s11033-023-08812-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Accepted: 09/11/2023] [Indexed: 10/13/2023]
Abstract
BACKGROUND Multiple sclerosis (MS) is a central nervous system disease known for immune-mediated demyelination, inflammatory, and neurodegeneration symptoms. Discovering molecular biomarkers to classify RRMS and SPMS patients, monitor the disease activity, and response to particular treatments is one area that has received notable attraction. MicroRNA (miRNA), a single-stranded non-coding RNA molecule, is a significant regulator of gene expression recruited in pathogenic mechanisms in diverse diseases, especially cancer and MS. Also, the relapsing-remitting features of MS exhibit that both inflammatory and anti-inflammatory cytokines are effective in the progression of the disease over time. METHODS AND RESULTS It was assessed the expression patterns of the genes (Drosha, Pasha (DGCR8), and Dicer ) encoding the critical enzymes in the processing steps of miRNA maturation and major pro-inflammatory and anti-inflammatory cytokines (IFN-α, IFN-β, and IL-6) in blood cells of 40 MS patients (two groups of 10 men and women in both clinical courses of RR and SPMS patients) in comparison with 20 healthy control group (10 males and 10 females). The highest transcription activity of Drosha was observed for RRMS patients (4.2 and 3.6-fold, respectively), and the expression ratio was down regulated in male and female patients with SPMS (3.9- and 3.1-fold, respectively). Considering the studied cytokines, the increase in expression ratio of IL-6 in SPMS patients and the decrease in transcript abundance of INF-α, and INF-β cytokines are consistent with the progression of the disease. CONCLUSIONS Our findings showed that the high and low transcriptional levels of the considered genes seem to be effective in the pathogenesis and progression of MS.
Collapse
Affiliation(s)
- Fatemeh Khakdan
- Department of Biology, Farzanegan Campus, Semnan University, Semnan, Iran
| | | | - Parisa Shahmoradipour
- Department of Biotechnology, Institute of Science, High Technology and Environmental Sciences, Graduate University of Advanced Technology, Kerman, Iran
| | | |
Collapse
|
10
|
Gao Y, Wang Y, Li M, Gao C. Bioinformatics analysis of potential common pathogenic mechanisms for systemic lupus erythematosus and acute myocardial infarction. Lupus 2023; 32:1296-1309. [PMID: 37800460 DOI: 10.1177/09612033231202659] [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] [Indexed: 10/07/2023]
Abstract
BACKGROUND Systemic lupus erythematosus (SLE) patients have a higher risk of acute myocardial infarction (AMI) compared to the general population. However, the underlying common mechanism of this association is not fully understood. This study aims to investigate the molecular mechanism of this complication. METHODS Gene expression profiles of SLE (GSE50772) and AMI (GSE66360) were obtained from the Gene Expression Omnibus (GEO) database. Common differentially expressed genes (DEGs) in SLE and AMI were identified, and functional annotation, protein-protein interaction (PPI) network analysis, module construction, and hub gene identification were performed. Additionally, transcription factor (TF)-gene regulatory network and TF-miRNA regulatory network were constructed for the hub genes. RESULTS 70 common DEGs (7 downregulated genes and 63 upregulated genes) were identified and were mostly enriched in signaling pathways such as the IL-17 signaling pathway, TNF signaling pathway, lipid metabolism, and atherosclerosis. Using cytoHubba, 12 significant hub genes were identified, including IL1B, TNF, FOS, CXCL8, JUN, PTGS2, FN1, EGR1, CXCL1, DUSP1, MMP9, and ZFP36. CONCLUSIONS This study reveals a common pathogenesis of SLE and AMI and provides new perspectives for further mechanism research. The identified common pathways and hub genes may have important clinical implications for the prevention and treatment of AMI in SLE patients.
Collapse
Affiliation(s)
- Yang Gao
- Department of Cardiology, Zhengzhou University People's Hospital, Henan Provincial People's Hospital, Fuwai Central China Cardiovascular Hospital, Zhengzhou, Henan, China
| | - Yunxia Wang
- Department of Radiology, Henan University People's Hospital, Henan Provincial People's Hospital, Zhengzhou, Henan, China
| | - Muwei Li
- Department of Cardiology, Zhengzhou University People's Hospital, Henan Provincial People's Hospital, Fuwai Central China Cardiovascular Hospital, Zhengzhou, Henan, China
| | - Chuanyu Gao
- Department of Cardiology, Zhengzhou University People's Hospital, Henan Provincial People's Hospital, Fuwai Central China Cardiovascular Hospital, Zhengzhou, Henan, China
| |
Collapse
|
11
|
Ferreira IA, Peixoto D, Losada AP, Quiroga MI, do Vale A, Costas B. Early innate immune responses in European sea bass ( Dicentrarchus labrax L.) following Tenacibaculum maritimum infection. Front Immunol 2023; 14:1254677. [PMID: 37731496 PMCID: PMC10507263 DOI: 10.3389/fimmu.2023.1254677] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Accepted: 08/15/2023] [Indexed: 09/22/2023] Open
Abstract
Introduction The marine aquaculture industry has been witnessing a worldwide emergence of tenacibaculosis, a poorly understood bacterial disease caused by Tenacibaculum maritimum that affects commercially important fish. So far, knowledge on the T. maritimum virulence mechanisms is scarce and the pathogen-host interaction operating in tenacibaculosis remain to be disclosed. This study aimed at contributing to a better understanding of this disease, by evaluating the early innate immune response triggered in European sea bass (Dicentrarchus labrax) by a bath-challenge with T. maritimum. Methods Groups of sea bass were bath-challenged with T. maritimum (challenged fish) or mock-challenged. Undisturbed fish were used as controls (time 0). Samples of blood, liver and mucosal organs (skin, gills and posterior-intestine) were collected at 0 h (control) and at 6, 24, 48 and 72 h post-challenge (n=12). Mucosal organs were used for analyzing the expression of immune-related genes by RT-qPCR, as well as blood samples for assessing haematological and innate humoral parameters and liver for oxidative stress assessment. Results An increased expression of il-1β, il8, mmp9 and hamp1 was detected in all mucosal organs of infected fish when compared with control and mock-challenged fish, suggesting a pro-inflammatory response against T. maritimum transversal to all organs. The faster induction of these pro-inflammatory genes was observed in the gills. Regarding the systemic response, challenged fish presented neutrophilia, monocytosis, signs of anemia, and a decrease of bactericidal and lysozyme activities in plasma. Almost no variations were observed regarding hepatic oxidative stress. Discussion/Conclusions The present study suggests that T. maritimum induces a local innate immune response upon bath infection not only in the skin of European sea bass, but also in the gills and posterior-intestine, likely triggered by the T. maritimum's capacity to adhere, colonize and damage these organs that can function as entry ways to bacteria, leading ultimately to the seen host's systemic response.
Collapse
Affiliation(s)
- Inês A. Ferreira
- Abel Salazar Institute of Biomedical Sciences (ICBAS), University of Porto, Porto, Portugal
- Interdisciplinary Centre of Marine and Environmental Research (CIIMAR), University of Porto, Porto, Portugal
- Fish Immunology and Vaccinology Group, IBMC-Instituto de Biologia Molecular e Celular, Universidade do Porto, Porto, Portugal
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
| | - Diogo Peixoto
- Abel Salazar Institute of Biomedical Sciences (ICBAS), University of Porto, Porto, Portugal
- Interdisciplinary Centre of Marine and Environmental Research (CIIMAR), University of Porto, Porto, Portugal
| | - Ana Paula Losada
- Departamento de Anatomía, Produción Animal e Ciencias Clínicas Veterinarias, Facultade de Veterinaria, Universidade de Santiago de Compostela, Lugo, Spain
| | - María Isabel Quiroga
- Departamento de Anatomía, Produción Animal e Ciencias Clínicas Veterinarias, Facultade de Veterinaria, Universidade de Santiago de Compostela, Lugo, Spain
| | - Ana do Vale
- Fish Immunology and Vaccinology Group, IBMC-Instituto de Biologia Molecular e Celular, Universidade do Porto, Porto, Portugal
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
| | - Benjamín Costas
- Abel Salazar Institute of Biomedical Sciences (ICBAS), University of Porto, Porto, Portugal
- Interdisciplinary Centre of Marine and Environmental Research (CIIMAR), University of Porto, Porto, Portugal
| |
Collapse
|
12
|
Mohsen Hammad DB, Abdulazeez Alhamad O, Mahdy obiad Khzal A, Mahdi Muslim Alameedy F. Molecular Characterisation of Blood Microbiome in Patients with Ankylosing Spondylitis and Healthy Controls. Med J Islam Repub Iran 2023; 37:84. [PMID: 38021395 PMCID: PMC10657266 DOI: 10.47176/mjiri.37.84] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Indexed: 12/01/2023] Open
Abstract
Background In human and animal studies, ankylosing spondylitis (AS) has been increasingly linked to changes in the microbial inhabitants in the human body (microbiome). These studies have primarily now concentrated on the microbial communities that live in the gastrointestinal tract. However, evidence suggests that various molecular techniques can be used to detect microbial DNA in blood circulation. This DNA might be an unknown reservoir of biomarkers with the potential to track alterations in the microbiomes of remote locations, such as the gut. To this end, we compared the presence and identity of microbial DNA in blood samples taken from ankylosing spondylitis patients to healthy control subjects by amplifying and sequencing the bacterial 16S rRNA variable region four. Methods The study's design is a case study based on the presence and identity of bacterial DNA in the blood of Ankylosing spondylitis (AS) patients (n = 10) and healthy control subjects (n = 10) was investigated by amplifying and sequencing the bacterial 16S rRNA gene. Blood concentrations of the cytokines TNF alpha, IL-17A, and IL-23 were determined by the Human Magnetic Luminex Screening, and data were analysed using an Unpaired T-test. Results Using PCR amplification, 8 of 10 AS patients (80%) and 8 of 10 healthy control samples (80%) had microbial 16S rRNA in their blood. At the phylum level, Proteobacteria (Control = 48.5%, AS = 52%), Firmicutes (Control = 27.8%, AS = 26.1%), Actinobacteria (Control = 15.4%, AS = 10.7%), and Bacteroidetes (Control = 6.5%, AS = 10%) dominated the blood microbiome. A two-tailed Mann-Whitney test found that Ankylosing Spondylitis was associated with significantly elevated Bacteroides (P < 0.05), Prevotella (P < 0.001), and Micrococcus (P < 0.01), and significantly reduced levels of Corynebacterium 1 (P < 0.001), Gemella (P < 0.01), and Alloprevotella (P < 0.05), compared to healthy controls. Additionally, it was shown that the presence of the Prevotella genus was highly positively correlated with higher levels of TNF-alpha (P < 0.05; r = 0.8) in AS patients' blood. Conclusion This article reveals that a blood microbiome exists in healthy individuals and identifies particular taxa modulated in disease. These blood-derived signatures indicate that this field needs more research and may be helpful as disease biomarkers.
Collapse
Affiliation(s)
| | - Omar Abdulazeez Alhamad
- Department of Biology, College of Education for Pure Science, University of
Mosul, Mosul, Iraq
| | | | | |
Collapse
|
13
|
Pasdaran A, Hassani B, Tavakoli A, Kozuharova E, Hamedi A. A Review of the Potential Benefits of Herbal Medicines, Small Molecules of Natural Sources, and Supplements for Health Promotion in Lupus Conditions. Life (Basel) 2023; 13:1589. [PMID: 37511964 PMCID: PMC10416186 DOI: 10.3390/life13071589] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Revised: 07/05/2023] [Accepted: 07/12/2023] [Indexed: 07/30/2023] Open
Abstract
The Latin word lupus, meaning wolf, was in the medical literature prior to the 1200s to describe skin lesions that devour flesh, and the resources available to physicians to help people were limited. The present text reviews the ethnobotanical and pharmacological aspects of medicinal plants and purified molecules from natural sources with efficacy against lupus conditions. Among these molecules are artemisinin and its derivatives, antroquinonol, baicalin, curcumin, emodin, mangiferin, salvianolic acid A, triptolide, the total glycosides of paeony (TGP), and other supplements such as fatty acids and vitamins. In addition, medicinal plants, herbal remedies, mushrooms, and fungi that have been investigated for their effects on different lupus conditions through clinical trials, in vivo, in vitro, or in silico studies are reviewed. A special emphasis was placed on clinical trials, active phytochemicals, and their mechanisms of action. This review can be helpful for researchers in designing new goal-oriented studies. It can also help practitioners gain insight into recent updates on supplements that might help patients suffering from lupus conditions.
Collapse
Affiliation(s)
- Ardalan Pasdaran
- Department of Pharmacognosy, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz 7146864685, Iran
- Medicinal Plants Processing Research Center, Shiraz University of Medical Sciences, Shiraz 7146864685, Iran
| | - Bahareh Hassani
- Student Research Committee, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz 7146864685, Iran
| | - Ali Tavakoli
- Research Center for Traditional Medicine and History of Medicine, Department of Persian Medicine, School of Medicine, Shiraz University of Medical Sciences, Shiraz 7134845794, Iran
| | - Ekaterina Kozuharova
- Department of Pharmacognosy, Faculty of Pharmacy, Medical University of Sofia, 1431 Sofia, Bulgaria
| | - Azadeh Hamedi
- Department of Pharmacognosy, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz 7146864685, Iran
- Medicinal Plants Processing Research Center, Shiraz University of Medical Sciences, Shiraz 7146864685, Iran
| |
Collapse
|
14
|
Winikajtis-Burzyńska A, Brzosko M, Przepiera-Będzak H. Increased Serum Interleukin 10 Levels Are Associated with Increased Disease Activity and Increased Risk of Anti-SS-A/Ro Antibody Positivity in Patients with Systemic Lupus Erythematosus. Biomolecules 2023; 13:974. [PMID: 37371554 PMCID: PMC10296207 DOI: 10.3390/biom13060974] [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: 04/30/2023] [Revised: 06/08/2023] [Accepted: 06/10/2023] [Indexed: 06/29/2023] Open
Abstract
Interleukin 10 (IL-10) plays a role in inflammation and cell-type responses. The anti-SS-A/Ro antibody contributes to leucopenia, and cutaneous and neonatal lupus. OBJECTIVES To evaluate the association between serum IL-10 levels and autoantibodies, disease activity and organ involvement in systemic lupus erythematosus (SLE) patients. PATIENTS AND METHODS We studied 200 SLE patients and 50 controls. We analyzed organ involvement, disease activity, serum IL-10 and interleukin-6 (IL-6) levels, and antinuclear and antiphospholipid antibody profiles. RESULTS Serum IL-10 and IL-6 levels were higher in SLE patients than in controls (all p < 0.00001). Serum IL-10 levels were positively correlated with IL-6 (p < 0.00001), CRP (p < 0.00001), fibrinogen (p = 0.003), and ESR (p < 0.00001), and negatively correlated with hemoglobin (p = 0.0004) and lymphocytes (p = 0.01). Serum IL-6 levels were positively correlated with CRP (p < 0.00001), fibrinogen (p = 0.001), and ESR (p < 0.00001); and negatively correlated with hemoglobin (p = 0.008) and lymphocytes (p = 0.03). Elevated serum IL-10 levels were associated with an increased risk of anti-SS-A/Ro antibody positivity (p = 0.03). Elevated serum IL-6 levels were associated with an increased risk of heart (p = 0.007) and lung (p = 0.04) involvement. CONCLUSIONS In SLE patients, increased serum IL-10 levels were associated with increased disease activity and risk of anti-SS-A/Ro antibody positivity.
Collapse
Affiliation(s)
- Agnieszka Winikajtis-Burzyńska
- Individual Laboratory for Rheumatologic Diagnostics, Pomeranian Medical University in Szczecin, Unii Lubelskiej 1, 71-252 Szczecin, Poland;
| | - Marek Brzosko
- Department of Rheumatology, Internal Medicine, Geriatrics and Clinical Immunology, Pomeranian Medical University in Szczecin, Unii Lubelskiej 1, 71-252 Szczecin, Poland;
| | - Hanna Przepiera-Będzak
- Department of Rheumatology, Internal Medicine, Geriatrics and Clinical Immunology, Pomeranian Medical University in Szczecin, Unii Lubelskiej 1, 71-252 Szczecin, Poland;
| |
Collapse
|
15
|
Sidheeque Hassan V, Hanifa M, Navik U, Bali A. Exogenous fetuin-A protects against sepsis-induced myocardial injury by inhibiting oxidative stress and inflammation in mice. Fundam Clin Pharmacol 2023; 37:607-617. [PMID: 36647295 DOI: 10.1111/fcp.12870] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Revised: 12/23/2022] [Accepted: 01/09/2023] [Indexed: 01/18/2023]
Abstract
Sepsis-induced myocardial injury is a consequence of septicemia and is one of the major causes of death in intensive care units. A serum glycoprotein called fetuin-A is secreted largely by the liver, tongue, placenta, and adipose tissue. Fetuin-A has a variety of biological and pharmacological properties. The anti-inflammatory and antioxidant glycoprotein fetuin-A has shown its efficacy in a number of inflammatory disorders including sepsis. However, its protective role against sepsis-induced myocardial injury remains elusive. The purpose of this work is to explore the role of fetuin-A in mouse models of myocardial injury brought on by cecal ligation and puncture (CLP). CLP significantly induced the myocardial injury assessed in terms of elevated myocardial markers (serum CK-MB, cTnI levels), inflammatory markers (IL-6, TNF-α) in the serum, and oxidative stress markers (increased MDA levels and decreased reduced glutathione) in heart tissue homogenate following 24 h of ligation and puncture. Further, hematoxylin and eosin (H&E) staining showed considerable histological alterations in the myocardial tissue of sepsis-developed mice. Interestingly, fetuin-A pretreatment (50 and 100 mg/kg) for 4 days before the CLP procedure significantly improved the myocardial injury and was evaluated in perspective of a reduction in the CK-MB, cTnI levels, IL-6, and TNF-α in sepsis-developed animals. Fetuin-A pretreatment significantly attenuated the oxidative stress and improved the myocardial morphology in a dose-dependent manner. The present study provides preliminary evidence that fetuin-A exerts protection against sepsis-induced cardiac dysfunction in vivo via suppression of inflammation and oxidative damage.
Collapse
Affiliation(s)
- V Sidheeque Hassan
- Department of Pharmacology, Central University of Punjab, Bathinda, India
| | - Mohd Hanifa
- Department of Pharmacology, Central University of Punjab, Bathinda, India
| | - Umashanker Navik
- Department of Pharmacology, Central University of Punjab, Bathinda, India
| | - Anjana Bali
- Department of Pharmacology, Central University of Punjab, Bathinda, India
| |
Collapse
|
16
|
Nikolakis D, Garantziotis P, Sentis G, Fanouriakis A, Bertsias G, Frangou E, Nikolopoulos D, Banos A, Boumpas DT. Restoration of aberrant gene expression of monocytes in systemic lupus erythematosus via a combined transcriptome-reversal and network-based drug repurposing strategy. BMC Genomics 2023; 24:207. [PMID: 37072752 PMCID: PMC10114456 DOI: 10.1186/s12864-023-09275-8] [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: 08/24/2022] [Accepted: 03/27/2023] [Indexed: 04/20/2023] Open
Abstract
BACKGROUND Monocytes -key regulators of the innate immune response- are actively involved in the pathogenesis of systemic lupus erythematosus (SLE). We sought to identify novel compounds that might serve as monocyte-directed targeted therapies in SLE. RESULTS We performed mRNA sequencing in monocytes from 15 patients with active SLE and 10 healthy individuals. Disease activity was assessed with the Systemic Lupus Erythematosus Disease Activity Index 2000 (SLEDAI-2 K). Leveraging the drug repurposing platforms iLINCS, CLUE and L1000CDS2, we identified perturbagens capable of reversing the SLE monocyte signature. We identified transcription factors and microRNAs (miRNAs) that regulate the transcriptome of SLE monocytes, using the TRRUST and miRWalk databases, respectively. A gene regulatory network, integrating implicated transcription factors and miRNAs was constructed, and drugs targeting central components of the network were retrieved from the DGIDb database. Inhibitors of the NF-κB pathway, compounds targeting the heat shock protein 90 (HSP90), as well as a small molecule disrupting the Pim-1/NFATc1/NLRP3 signaling axis were predicted to efficiently counteract the aberrant monocyte gene signature in SLE. An additional analysis was conducted, to enhance the specificity of our drug repurposing approach on monocytes, using the iLINCS, CLUE and L1000CDS2 platforms on publicly available datasets from circulating B-lymphocytes, CD4+ and CD8+ T-cells, derived from SLE patients. Through this approach we identified, small molecule compounds, that could potentially affect more selectively the transcriptome of SLE monocytes, such as, certain NF-κB pathway inhibitors, Pim-1 and SYK kinase inhibitors. Furthermore, according to our network-based drug repurposing approach, an IL-12/23 inhibitor and an EGFR inhibitor may represent potential drug candidates in SLE. CONCLUSIONS Application of two independent - a transcriptome-reversal and a network-based -drug repurposing strategies uncovered novel agents that might remedy transcriptional disturbances of monocytes in SLE.
Collapse
Affiliation(s)
- Dimitrios Nikolakis
- Amsterdam Institute for Gastroenterology Endocrinology and Metabolism, Department of Gastroenterology, Amsterdam UMC, Academic Medical Center, University of Amsterdam, Meibergdreef 9, Amsterdam, 1105 AZ, The Netherlands
- Department of Rheumatology and Clinical Immunology, Amsterdam Rheumatology & Immunology Center (ARC), Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
- Amsterdam Institute for Infection & Immunity, Department of Experimental Immunology, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
- Onassis Foundation, Athens, Greece
| | - Panagiotis Garantziotis
- Laboratory of Autoimmunity and Inflammation, Center for Clinical, Experimental Surgery and Translational Research, Biomedical Research Foundation of the Academy of Athens, Athens, Greece
- Department Rheumatology and Immunology, Hannover Medical School, Hannover, Germany
| | - George Sentis
- Laboratory of Autoimmunity and Inflammation, Center for Clinical, Experimental Surgery and Translational Research, Biomedical Research Foundation of the Academy of Athens, Athens, Greece
| | - Antonis Fanouriakis
- Rheumatology and Clinical Immunology Unit, Department of Internal Medicine, Attikon University Hospital, Athens, 4th, Greece
- Department of Propaedeutic Internal Medicine, "Laiko" General Hospital, Athens, Greece
- Joint Academic Rheumatology Program, National and Kapodistrian University of Athens Medical School, Athens, Greece
| | - George Bertsias
- Department of Rheumatology and Clinical Immunology, Medical School, University Hospital of Heraklion, University of Crete, Heraklion, Greece
- Institute of Molecular Biology and Biotechnology-FORTH, Heraklion, Greece
| | - Eleni Frangou
- Laboratory of Autoimmunity and Inflammation, Center for Clinical, Experimental Surgery and Translational Research, Biomedical Research Foundation of the Academy of Athens, Athens, Greece
- Department of Nephrology, Limassol General Hospital, Limassol, Cyprus
- Medical School, University of Nicosia, Nicosia, Cyprus
| | - Dionysis Nikolopoulos
- Laboratory of Autoimmunity and Inflammation, Center for Clinical, Experimental Surgery and Translational Research, Biomedical Research Foundation of the Academy of Athens, Athens, Greece
- Rheumatology and Clinical Immunology Unit, Department of Internal Medicine, Attikon University Hospital, Athens, 4th, Greece
| | - Aggelos Banos
- Laboratory of Autoimmunity and Inflammation, Center for Clinical, Experimental Surgery and Translational Research, Biomedical Research Foundation of the Academy of Athens, Athens, Greece
| | - Dimitrios T Boumpas
- Laboratory of Autoimmunity and Inflammation, Center for Clinical, Experimental Surgery and Translational Research, Biomedical Research Foundation of the Academy of Athens, Athens, Greece.
- Rheumatology and Clinical Immunology Unit, Department of Internal Medicine, Attikon University Hospital, Athens, 4th, Greece.
- Joint Academic Rheumatology Program, National and Kapodistrian University of Athens Medical School, Athens, Greece.
| |
Collapse
|
17
|
Caruso G, Di Pietro L, Cardaci V, Maugeri S, Caraci F. The therapeutic potential of carnosine: Focus on cellular and molecular mechanisms. CURRENT RESEARCH IN PHARMACOLOGY AND DRUG DISCOVERY 2023. [DOI: 10.1016/j.crphar.2023.100153] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/08/2023] Open
|
18
|
Ni FF, Liu GL, Jia SL, Li CR, Gao XJ. Effects of the mTOR Pathway on the Balance of Th2/Treg Cells in Children with Idiopathic Nephrotic Syndrome. Indian J Nephrol 2023; 33:93-100. [PMID: 37234433 PMCID: PMC10208544 DOI: 10.4103/ijn.ijn_521_21] [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: 12/15/2021] [Revised: 05/19/2022] [Accepted: 05/29/2022] [Indexed: 05/28/2023] Open
Abstract
Introduction Immune dysfunction contributes to the progression of idiopathic nephrotic syndrome (INS), but the details of the pathogenesis of progression remain unknown. This study of children with INS investigated the relationship of activation of the mechanistic target of rapamycin (mTOR) pathway (PI3K/AKT/mTOR/p70S6K) with the levels of T helper 2/regulatory T (Th2/Treg) cells. Materials and Methods Twenty children with active INS (before steroid treatment), 20 children with remitting INS (INS-R, after steroid treatment), and 20 healthy control children (Ctrl) were enrolled. The levels of Th2/Treg cells in their peripheral circulatory systems were measured using flow cytometry, and the concentration of interleukin (IL)-4 was determined using a cytometric bead array (CBA). The levels of PI3K, AKT, mTOR, p70S6K, and transcription factors associated with Th2/Treg cells were measured using real-time polymerase chain reaction. Results The INS group had a greater proportion of circulating Th2 cells; level of IL-4 protein; and levels of GATA, PI3K, AKT, mTOR, and p70S6K mRNAs than the Ctrl group (all P < 0.05), but a lower proportion of circulating Tregs and expression of Foxp3 (both P < 0.05). Patients in the INS-R group had normalization of these markers (all P < 0.05). Patients in the INS group had negative correlation in the percentage of Treg cells with Th2 cells and with IL-4 level and a negative correlation in the levels of GATA3 and Foxp3 mRNAs. Conclusions Patients with active INS had an imbalance of Th2/Treg cells, which might result from the aberrant signaling of the mTOR pathway (PI3K/AKT/mTOR/p70S6K).
Collapse
Affiliation(s)
- Fen Fen Ni
- Department of Nephrology, Shenzhen Children’s Hospital, Shenzhen, China
| | - Guang Lei Liu
- Department of Pediatrics, The Fifth Affiliated (Zhuhai) Hospital of Zunyi Medical University, Zhuhai, China
| | - Shi Lei Jia
- Department of Nephrology, Shenzhen Children’s Hospital, Shenzhen, China
| | - Cheng Rong Li
- Department of Nephrology, Shenzhen Children’s Hospital, Shenzhen, China
| | - Xiao Jie Gao
- Department of Nephrology, Shenzhen Children’s Hospital, Shenzhen, China
| |
Collapse
|
19
|
Green tea silver nanoparticles improve physiological motor and cognitive function in BALB/c mice during inflammation. Heliyon 2023; 9:e13922. [PMID: 36873543 PMCID: PMC9982668 DOI: 10.1016/j.heliyon.2023.e13922] [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: 07/12/2022] [Revised: 02/08/2023] [Accepted: 02/16/2023] [Indexed: 02/24/2023] Open
Abstract
Information on the basic changes associated with green tea small molecules in acute inflammation is deficient. The purpose of the study was to characterize and establish the effects of green tea silver nanoparticles (AgNPs) following inflammation in BALB/c male mice. In this study, green tea silver nitrate nanoparticles were characterized and the extract were made up to constitute high (100%), medium (10%), and low (1%) concentrations for administration. Acute inflammation was induced in groups I-V of the experimental rodents by injecting 0.5 ml/kg of fresh egg albumin on the subplantar surface of the right hind paw and animals were monitored for 36 h. Group I-III were administered 100%, 10%, 1% green tea nanoparticles extract while group IV was given diclofenac. Group V was the positive control while group VI was the negative control that received the vehicle. Paw edema was measured at a 2 h interval for 3 days, while the pain was assessed by measuring the locomotion activity using the voluntary wheel running and the anxiety-like behavior. Hypersensitivity was measured through the temperature sensation experiment and a non-linear regression analysis was done. Here, synthesized green tea AgNPs registered an absorbance band at 460 nm, phytochemicals due to presence of organic functional groups of O[bond, double bond]C[bond, double bond]O of oxycarbons, of C[bond, double bond]C of a conjugate alkene, C[bond, double bond]O of a stretching bond of a secondary alcohol. The silver green tea nanoparticles were spherical, covered by a slimy layer, capped and stable. Green tea AgNPs significantly decreased temperature hypersensitivity in BALB/c male mice and this demonstrated their protective effects. Low concentrations of green tea nanoparticles inhibited edema thus mimicking effects of diclofenac, however, the percentage of inhibition was highest in medium and high silver-tea nanoparticles concentrations demonstration the importance of concentration in therapeutics. Anxiety was lowest in BALB/c male mice treated with high concentrations of silver green tea nanoparticles, and this led to increased locomotory activity in mice. Green tea AgNPs have strong anti-inflammatory effects at high concentrations. Concentrations of green tea AgNPs modulated basic sensory and motor behaviors in BALB/c male mice demonstrating their importance in complementary and integrative medical practice.
Collapse
|
20
|
Patnaik SS, Kotipalli R, Jerald MK, Muralidharan K. Combination treatment of recombinant growth differentiation factor-9 and Cetrorelix improves gestational origin of the polycystic ovarian syndrome in female rats. Life Sci 2023; 321:121638. [PMID: 37001808 DOI: 10.1016/j.lfs.2023.121638] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Revised: 03/16/2023] [Accepted: 03/24/2023] [Indexed: 03/31/2023]
Abstract
AIMS Polycystic ovary syndrome (PCOS) is a hyper-androgenic endocrinopathy prevalent in premenopausal women with no cure available. The current study aimed to investigate the therapeutic effect of recombinant GDF-9 and Cetrorelix on the gestational origin of dehydroepiandrosterone (DHEA) induced PCOS in postnatal pups' delivered to rat dams. MAIN METHODS The body weight measurement, blood and serum analysis for glucose tolerance, lipid profile, liver enzymes, sex hormones (Testosterone, Estradiol, and Progesterone), estrus cyclicity assessment, histological staining of ovary and liver, molecular markers expressions of pro-inflammatory by qRT-PCR and immuno-histochemistry technique for folliculogenesis genes and histological staining studies of liver and ovary were done. KEY FINDINGS The combinational treatment was found to normalize the biochemical parameters and reduction in the estrus irregularity by altering the sex hormones as well as the glucose metabolism and insulin resistance via HOMA-IR value. Further, molecular markers expression confirmed the pro-inflammatory (IL-1β, TNF-α, and IL-6) and folliculogenesis (GDF-9, BMPR2, and TGF-βR1) genes associated with PCOS were improved by combinational therapy. SIGNIFICANCE In conclusion, rGDF-9 could be a potential therapeutic agent in combination with Cetrorelix as a better treatment regime for metabolic and reproductive phenotypes in PCOS. However, the effect of rGDF-9 on infertility-associated phenotypes in PCOS needs further evaluation.
Collapse
|
21
|
Ochando J, Mulder WJM, Madsen JC, Netea MG, Duivenvoorden R. Trained immunity - basic concepts and contributions to immunopathology. Nat Rev Nephrol 2023; 19:23-37. [PMID: 36253509 PMCID: PMC9575643 DOI: 10.1038/s41581-022-00633-5] [Citation(s) in RCA: 103] [Impact Index Per Article: 103.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/13/2022] [Indexed: 02/08/2023]
Abstract
Trained immunity is a functional state of the innate immune response and is characterized by long-term epigenetic reprogramming of innate immune cells. This concept originated in the field of infectious diseases - training of innate immune cells, such as monocytes, macrophages and/or natural killer cells, by infection or vaccination enhances immune responses against microbial pathogens after restimulation. Although initially reported in circulating monocytes and tissue macrophages (termed peripheral trained immunity), subsequent findings indicate that immune progenitor cells in the bone marrow can also be trained (that is, central trained immunity), which explains the long-term innate immunity-mediated protective effects of vaccination against heterologous infections. Although trained immunity is beneficial against infections, its inappropriate induction by endogenous stimuli can also lead to aberrant inflammation. For example, in systemic lupus erythematosus and systemic sclerosis, trained immunity might contribute to inflammatory activity, which promotes disease progression. In organ transplantation, trained immunity has been associated with acute rejection and suppression of trained immunity prolonged allograft survival. This novel concept provides a better understanding of the involvement of the innate immune response in different pathological conditions, and provides a new framework for the development of therapies and treatment strategies that target epigenetic and metabolic pathways of the innate immune system.
Collapse
Affiliation(s)
- Jordi Ochando
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA. .,Transplant Immunology Unit, National Center of Microbiology, Instituto de Salud Carlos III, Madrid, Spain.
| | - Willem J. M. Mulder
- grid.6852.90000 0004 0398 8763Laboratory of Chemical Biology, Department of Biomedical Engineering and Institute for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, The Netherlands ,grid.59734.3c0000 0001 0670 2351Biomedical Engineering and Imaging Institute, Department of Radiology, Icahn School of Medicine at Mount Sinai, New York, NY USA
| | - Joren C. Madsen
- grid.32224.350000 0004 0386 9924Center for Transplantation Sciences, Department of Surgery, Massachusetts General Hospital, Boston, MA USA ,grid.32224.350000 0004 0386 9924Division of Cardiac Surgery, Department of Surgery, Massachusetts General Hospital, Boston, MA USA
| | - Mihai G. Netea
- grid.10417.330000 0004 0444 9382Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, The Netherlands ,grid.10388.320000 0001 2240 3300Department for Genomics & Immunoregulation, Life and Medical Sciences Institute (LIMES), University of Bonn, Bonn, Germany
| | - Raphaël Duivenvoorden
- Biomedical Engineering and Imaging Institute, Department of Radiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA. .,Department of Nephrology, Radboud University Medical Center, Nijmegen, The Netherlands.
| |
Collapse
|
22
|
El-Akhras BA, Talaat RM, El-Masry SA, Bassyouni IH, El-Sayed IH, Ali YBM. Crosstalk between miR-146a and pro-inflammatory cytokines in patients with systemic lupus erythematosus. Int J Immunopathol Pharmacol 2023; 37:3946320231154998. [PMID: 36740569 PMCID: PMC9903020 DOI: 10.1177/03946320231154998] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
microRNA-146a (miR-146a) plays an essential role in immune anomalies and organ injury of systemic lupus erythematosus (SLE) by regulating the disease's inflammation and complications. Here, we analyzed the expression of miR-146a in SLE and a panel of pro-inflammatory cytokines (IL-1, IL-6, IL-8, IL-17, and TNF-α). Association between all measured parameters and the disease's clinical manifestation and response to treatment was monitored. Our study populations were 113 SLE patients and 104 healthy volunteers. miR-146a expression in peripheral blood mononuclear cells (PBMCs) was measured by quantitative real-time PCR (RT-qPCR). The content of the plasma cytokines (IL-1β, IL-6, IL-8, IL-17, and TNF-α) was detected by enzyme-linked immunosorbent assay (ELISA). Compared with healthy controls, miR-146a expression was significantly increased (p < 0.05) in lupus patients. The analysis of the receiver operator characteristic curve (ROC) of miR-146a showed 91% sensitivity and 70% specificity. IL-1β, IL-6, and IL-17 cytokines were significantly increased (p < 0.001), while IL-8 and TNF-α were significantly decreased (p < 0.001) in SLE patients against controls. The expression of miR-146a and TNF-α was upregulated considerably in SLE patients with severe disease activity. miR-146a expression was positively correlated with IL-6. Our results pointed to the elevation of miR-146a as a trade marker of SLE patients. Reduction of IL-8 and TNF-α in combination with an elevation of IL-1β, IL-6, and IL-17 might refer to miR-146a's dual effect in controlling inflammation in lupus. Although we shed some light on the role of miR-146a in SLE, further study is recommended to improve our results.
Collapse
Affiliation(s)
- Basima A El-Akhras
- Molecular Biology Department, Genetic Engineering and Biotechnology Research Institute, Sadat City University, Egypt
| | - Roba M Talaat
- Molecular Biology Department, Genetic Engineering and Biotechnology Research Institute, Sadat City University, Egypt
| | - Samir A El-Masry
- Molecular Biology Department, Genetic Engineering and Biotechnology Research Institute, Sadat City University, Egypt
| | - Iman H Bassyouni
- Rheumatology and Rehabilitation Department, Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Ibrahim H El-Sayed
- Biochemistry Department, Faculty of Science, Kafr El-Sheikh University, Egypt
| | - Yasser BM Ali
- Molecular Biology Department, Genetic Engineering and Biotechnology Research Institute, Sadat City University, Egypt
| |
Collapse
|
23
|
Mora VP, Loaiza RA, Soto JA, Bohmwald K, Kalergis AM. Involvement of trained immunity during autoimmune responses. J Autoimmun 2022:102956. [DOI: 10.1016/j.jaut.2022.102956] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Accepted: 11/14/2022] [Indexed: 12/23/2022]
|
24
|
Melamud MM, Ermakov EA, Boiko AS, Kamaeva DA, Sizikov AE, Ivanova SA, Baulina NM, Favorova OO, Nevinsky GA, Buneva VN. Multiplex Analysis of Serum Cytokine Profiles in Systemic Lupus Erythematosus and Multiple Sclerosis. Int J Mol Sci 2022; 23:ijms232213829. [PMID: 36430309 PMCID: PMC9695219 DOI: 10.3390/ijms232213829] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Revised: 10/27/2022] [Accepted: 11/08/2022] [Indexed: 11/12/2022] Open
Abstract
Changes in cytokine profiles and cytokine networks are known to be a hallmark of autoimmune diseases, including systemic lupus erythematosus (SLE) and multiple sclerosis (MS). However, cytokine profiles research studies are usually based on the analysis of a small number of cytokines and give conflicting results. In this work, we analyzed cytokine profiles of 41 analytes in patients with SLE and MS compared with healthy donors using multiplex immunoassay. The SLE group included treated patients, while the MS patients were drug-free. Levels of 11 cytokines, IL-1b, IL-1RA, IL-6, IL-9, IL-10, IL-15, MCP-1/CCL2, Fractalkine/CX3CL1, MIP-1a/CCL3, MIP-1b/CCL4, and TNFa, were increased, but sCD40L, PDGF-AA, and MDC/CCL22 levels were decreased in SLE patients. Thus, changes in the cytokine profile in SLE have been associated with the dysregulation of interleukins, TNF superfamily members, and chemokines. In the case of MS, levels of 10 cytokines, sCD40L, CCL2, CCL3, CCL22, PDGF-AA, PDGF-AB/BB, EGF, IL-8, TGF-a, and VEGF, decreased significantly compared to the control group. Therefore, cytokine network dysregulation in MS is characterized by abnormal levels of growth factors and chemokines. Cross-disorder analysis of cytokine levels in MS and SLE showed significant differences between 22 cytokines. Protein interaction network analysis showed that all significantly altered cytokines in both SLE and MS are functionally interconnected. Thus, MS and SLE may be associated with impaired functional relationships in the cytokine network. A cytokine correlation networks analysis revealed changes in correlation clusters in SLE and MS. These data expand the understanding of abnormal regulatory interactions in cytokine profiles associated with autoimmune diseases.
Collapse
Affiliation(s)
- Mark M. Melamud
- Institute of Chemical Biology and Fundamental Medicine, Siberian Branch of the Russian Academy of Sciences, 630090 Novosibirsk, Russia
| | - Evgeny A. Ermakov
- Institute of Chemical Biology and Fundamental Medicine, Siberian Branch of the Russian Academy of Sciences, 630090 Novosibirsk, Russia
| | - Anastasiia S. Boiko
- Mental Health Research Institute, Tomsk National Research Medical Center of the Russian Academy of Sciences, 634014 Tomsk, Russia
| | - Daria A. Kamaeva
- Mental Health Research Institute, Tomsk National Research Medical Center of the Russian Academy of Sciences, 634014 Tomsk, Russia
| | - Alexey E. Sizikov
- Institute of Clinical Immunology, Siberian Branch of the Russian Academy of Sciences, 630099 Novosibirsk, Russia
| | - Svetlana A. Ivanova
- Mental Health Research Institute, Tomsk National Research Medical Center of the Russian Academy of Sciences, 634014 Tomsk, Russia
| | - Natalia M. Baulina
- Pirogov Russian National Research Medical University, 117997 Moscow, Russia
| | - Olga O. Favorova
- Pirogov Russian National Research Medical University, 117997 Moscow, Russia
| | - Georgy A. Nevinsky
- Institute of Chemical Biology and Fundamental Medicine, Siberian Branch of the Russian Academy of Sciences, 630090 Novosibirsk, Russia
| | - Valentina N. Buneva
- Institute of Chemical Biology and Fundamental Medicine, Siberian Branch of the Russian Academy of Sciences, 630090 Novosibirsk, Russia
- Correspondence: ; Tel.: +7-383-363-51-27
| |
Collapse
|
25
|
Hou G, Zhou T, Xu N, Yin Z, Zhu X, Zhang Y, Cui Y, Ma J, Tang Y, Cheng Z, Shen Y, Chen Y, Zou LH, Wang YF, Yin Z, Guo Y, Ding H, Ye Z, Shen N. Integrative Functional Genomics Identifies Systemic Lupus Erythematosus Causal Genetic Variant in the IRF5 Risk Locus. Arthritis Rheumatol 2022; 75:574-585. [PMID: 36245280 DOI: 10.1002/art.42390] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Revised: 09/14/2022] [Accepted: 10/04/2022] [Indexed: 11/09/2022]
Abstract
OBJECTIVE IRF5 plays a crucial role in the development of lupus. Genome-wide association studies have identified several systemic lupus erythematosus (SLE) risk single-nucleotide polymorphisms (SNPs) enriched in the IRF5 locus. However, no comprehensive genome editing-based functional analysis exists to establish a direct link between these variants and altered IRF5 expression, particularly for enhancer variants. This study was undertaken to dissect the regulatory function and mechanisms of SLE IRF5 enhancer risk variants and to explore the utilization of clustered regularly interspaced short palindromic repeat interference (CRISPRi) to regulate the expression of disease risk gene to intervene in the disease. METHODS Epigenomic profiles and expression quantitative trait locus analysis were applied to prioritize putative functional variants in the IRF5 locus. CRISPR-mediated deletion, activation, and interference were performed to investigate the genetic function of rs4728142. Allele-specific chromatin immunoprecipitation-quantitative polymerase chain reaction and allele-specific formaldehyde-assisted isolation of regulatory element-quantitative polymerase chain reaction were used to decipher the mechanism of alleles differentially regulating IRF5 expression. The CRISPRi approach was used to evaluate the intervention effect in monocytes from SLE patients. RESULTS SLE risk SNP rs4728142 was located in an enhancer region, indicating a disease-related regulatory function, and risk allele rs4728142-A was closely associated with increased IRF5 expression. We demonstrated that an rs4728142-containing region could act as an enhancer to regulate the expression of IRF5. Moreover, rs4728142 affected the binding affinity of zinc finger and BTB domain-containing protein 3 (ZBTB3), a transcription factor involved in regulation. Furthermore, in monocytes from SLE patients, CRISPR-based interference with the regulation of this enhancer attenuated the production of disease-associated cytokines. CONCLUSION These results demonstrate that the rs4728142-A allele increases the SLE risk by affecting ZBTB3 binding, chromatin status, and regulating IRF5 expression, establishing a biologic link between genetic variation and lupus pathogenesis.
Collapse
Affiliation(s)
- Guojun Hou
- Shanghai Institute of Rheumatology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China, Shenzhen Futian Hospital for Rheumatic Diseases, Shenzhen, China, and State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Tian Zhou
- Shanghai Institute of Rheumatology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ning Xu
- Shanghai Institute of Rheumatology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhihua Yin
- Shenzhen Futian Hospital for Rheumatic Diseases, and Joint Research Laboratory for Rheumatology of Shenzhen University Health Science Center and Shenzhen Futian Hospital for Rheumatic Diseases, Shenzhen, China
| | - Xinyi Zhu
- Shanghai Institute of Rheumatology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yutong Zhang
- Shanghai Institute of Rheumatology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yange Cui
- Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Jianyang Ma
- Shanghai Institute of Rheumatology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yuanjia Tang
- Shanghai Institute of Rheumatology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhaorui Cheng
- Shanghai Institute of Rheumatology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yiwei Shen
- Shanghai Institute of Rheumatology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yashuo Chen
- Shenzhen Futian Hospital for Rheumatic Diseases, and Joint Research Laboratory for Rheumatology of Shenzhen University Health Science Center and Shenzhen Futian Hospital for Rheumatic Diseases, Shenzhen, China
| | - Ling-Hua Zou
- Shenzhen Futian Hospital for Rheumatic Diseases, and Joint Research Laboratory for Rheumatology of Shenzhen University Health Science Center and Shenzhen Futian Hospital for Rheumatic Diseases, Shenzhen, China
| | - Yong-Fei Wang
- School of Life and Health Sciences, School of Medicine, and Warshel Institute for Computational Biology, The Chinese University of Hong Kong, Shenzhen, Guangdong, China
| | - Zihang Yin
- Sheng Yushou Center of Cell Biology and Immunology, Joint International Research Laboratory of Metabolic & Developmental Sciences, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, China
| | - Ya Guo
- Sheng Yushou Center of Cell Biology and Immunology, Joint International Research Laboratory of Metabolic & Developmental Sciences, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, China
| | - Huihua Ding
- Shanghai Institute of Rheumatology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhizhong Ye
- Shenzhen Futian Hospital for Rheumatic Diseases, and Joint Research Laboratory for Rheumatology of Shenzhen University Health Science Center and Shenzhen Futian Hospital for Rheumatic Diseases, Shenzhen, China
| | - Nan Shen
- Shanghai Institute of Rheumatology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China, Shenzhen Futian Hospital for Rheumatic Diseases, Shenzhen, China, State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China, Center for Autoimmune Genomics and Etiology, Division of Human Genetics, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, and Department of Pediatrics, University of Cincinnati, Cincinnati, Ohio
| |
Collapse
|
26
|
The Comprehensive Analysis of Hub Gene ARRB2 in Prostate Cancer. DISEASE MARKERS 2022; 2022:8518378. [PMID: 36284990 PMCID: PMC9588343 DOI: 10.1155/2022/8518378] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Revised: 08/23/2022] [Accepted: 09/06/2022] [Indexed: 12/01/2022]
Abstract
Methods The differential expressed genes (DEGs) were screened from the gene expression profile GSE30994 related to PRAD and then analyzed by protein-protein interaction (PPI) to screen the hub gene. Subsequently, the relation between hub gene and pan cancers, PRAD prognosis, and immunotherapy was analyzed. Besides, the effects of hub gene on the growth and metastasis of PRAD cell lines and inflammatory factors (IFs) were detected by functional experiments. Results 276 upregulated and 1,861 downregulated DEGs were analyzed from GSE30994 gene expression profiles. Through enrichment analysis, it was found that upregulated DEGs were significantly enriched in nitric oxide-mediated signal transduction, insulin signaling pathway, etc. Through PPI networks, ARRB2 was determined as the hub gene that was highly expressed in pan cancers, including PRAD, and contributed to poor prognosis of PRAD patients. Immunoassay showed that ARRB2 was associated with B cells, NK cells, endothelial cells, etc. and also connected with tumor-infiltrating lymphocytes (TILs). Next, the signature model analysis revealed that ARRB2 had a clinical value in predicting PRAD prognosis. In functional experiments, ARRB2 was highly expressed in PRAD cell lines, promoted PRAD cell growth and metastasis, and positively associated with IFs. Conclusion ARRB2 has a good prognostic ability in PRAD, and it could be a potential target of PRAD immunotherapy, which offers new directions for PRAD research.
Collapse
|
27
|
Liu M, Zhang H, Xie Z, Huang Y, Sun G, Qi D, Furey A, Randell EW, Rahman P, Zhai G. Glutathione, polyamine, and lysophosphatidylcholine synthesis pathways are associated with circulating pro-inflammatory cytokines. Metabolomics 2022; 18:76. [PMID: 36180605 DOI: 10.1007/s11306-022-01932-5] [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: 06/01/2022] [Accepted: 08/29/2022] [Indexed: 10/14/2022]
Abstract
INTRODUCTION Pro-inflammatory cytokines are responsible for initiating an effective defense against exogenous pathogens, and their regulation has a vital role in maintaining physiological homeostasis. The involvement of pro-inflammatory cytokines in pathological conditions have been explored in great detail, however, studies investigating metabolic pathways associated with these cytokines under normal homeostatic conditions are scarce. OBJECTIVES The aim of the current study was to identify metabolites and metabolic pathways associated with circulating pro-inflammatory cytokines under homeostatic conditions using a metabolomics approach. METHODS The study participants (n = 133) were derived from the Newfoundland Osteoarthritis Study (NFOAS) and the Complex Diseases in the Newfoundland population: Environment and Genetics (CODING) study. Plasma concentrations of cytokines including tumor necrosis factor-alpha (TNF-α), interleukin-6 (IL-6), interleukin-1 beta (IL-1β), and macrophage migration inhibitory factor (MIF) were assessed by enzyme-linked immunosorbent assay. Targeted metabolomic profiling on fasting plasma samples was performed using Biocrates MxP® Quant 500 kit which measures a total of 630 metabolites. Associations between natural log-transformed metabolite concentrations and metabolite sums/ratios and cytokine levels were assessed using linear regression with adjustment for age, sex, body mass index (BMI), and osteoarthritis status. RESULTS Seven metabolites and 11 metabolite sums/ratios were found to be significantly associated with TNF-α, IL-1β, and MIF (all p ≤ 5.13 × 10- 5) after controlling multiple testing with Bonferroni method, indicating the association between glutathione (GSH), polyamine, and lysophosphatidylcholine (lysoPC) synthesis pathways and these pro-inflammatory cytokines. CONCLUSION GSH, polyamine, and lysoPC synthesis pathways were positively associated with circulating TNF-α, IL-1β, and MIF levels under homeostatic conditions.
Collapse
Affiliation(s)
- Ming Liu
- Division of Biomedical Sciences (Genetics), Faculty of Medicine, Memorial University of Newfoundland, St. John's, Canada
| | - Hongwei Zhang
- Discipline of Medicine, Faculty of Medicine, Memorial University of Newfoundland, St. John's, Canada
| | - Zikun Xie
- Xiangya Hospital, Central South University, Changsha, China
| | - Yiheng Huang
- College of Pharmacy, University of Manitoba, Winnipeg, Canada
| | - Guang Sun
- Discipline of Medicine, Faculty of Medicine, Memorial University of Newfoundland, St. John's, Canada
| | - Dake Qi
- College of Pharmacy, University of Manitoba, Winnipeg, Canada
| | - Andrew Furey
- Discipline of Surgery, Faculty of Medicine, Memorial University of Newfoundland and Office of the Premier, Government of Newfoundland and Labrador, St. John's, Canada
| | - Edward W Randell
- Discipline of Laboratory Medicine, Faculty of Medicine, Memorial University of Newfoundland, St. John's, Canada
| | - Proton Rahman
- Discipline of Medicine, Faculty of Medicine, Memorial University of Newfoundland, St. John's, Canada
| | - Guangju Zhai
- Division of Biomedical Sciences (Genetics), Faculty of Medicine, Memorial University of Newfoundland, St. John's, Canada.
| |
Collapse
|
28
|
Functional and Structural Impact of Deleterious Missense Single Nucleotide Polymorphisms in the NR3C1, CYP3A5, and TNF-α Genes: An In Silico Analysis. Biomolecules 2022; 12:biom12091307. [PMID: 36139147 PMCID: PMC9496109 DOI: 10.3390/biom12091307] [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: 07/06/2022] [Revised: 08/24/2022] [Accepted: 08/31/2022] [Indexed: 11/16/2022] Open
Abstract
Human diseases are generally influenced by SNPs (single nucleotide polymorphisms). The mutations in amino acid residues generated by deleterious SNPs contribute to the structural and functional diversity of the encoded protein. Tumor necrosis factor-α (TNF-α), Glucocorticoid receptor gene (NR3C1), and Cytochrome P450 3A5 (CYP3A5) play a key role in glucocorticoid resistance susceptibility in humans. Possible causative mutations could be used as therapeutic targets and diagnostic markers for glucocorticoid resistance. This study evaluated the missense SNPs of TNF-α, NR3C1, and CYP3A5 to predict their impact on amino acid changes, protein interaction, and functional stability. The protein sequence of dbSNP was obtained and used online in silico method to screen deleterious mutants for the in silico analysis. In the coding regions of TNF-α, NR3C1, and CYP3A5, 14 deleterious mutations were discovered. The protein functional and stability changes in the amino acid between native and mutant energy were identified by analyzing the changes in the hydrogen bonding of these mutants from native, which were all measured using Swiss PDB and PyMOL. F446S and R439K had the highest root-mean-square deviation (RMSD) values among the 14 deleterious mutants. Additionally, the conserved region of amino acid protein interaction was analyzed. This study could aid in the discovery of new detrimental mutations in TNF-α, NR3C1, and CYP3A5, as well as the development of long-term therapy for corticosteroid resistance in several inflammatory diseases. However, more research into the deleterious mutations of the TNF-α, NR3C1, and CYP3A5 genes is needed to determine their role in corticosteroid resistance.
Collapse
|
29
|
Shi Z, Zhang YP, Hong D, Qiu X, Zheng L, Bian L, Hu F, Chen L, Xiong H, Yang Q, Jiang S, Tan G, Wang L. Anti-galectin-3 antibodies induce skin vascular inflammation via promoting local production of IL-1β in systemic lupus erythematosus. Int Immunopharmacol 2022; 112:109197. [PMID: 36058031 DOI: 10.1016/j.intimp.2022.109197] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Revised: 08/05/2022] [Accepted: 08/23/2022] [Indexed: 11/05/2022]
Abstract
Vascular inflammation could occur in all organs and tissues in patients with systematic lupus erythematosus (SLE), of which skin is the most frequent one. Our previous research identified anti-galectin-3 (Gal3) antibodies (Abs) as an important mediator of lupus cutaneous vasculopathy. Herein, we showed that anti-Gal3 Abs dysregulated the function of vascular endothelial cells with higher transcript levels of IL-1β and increased expression of mature IL-1β. The enhanced production of IL-1β secreted by endothelial cells was dependent on NLRP3 inflammasome. Intradermal injection of anti-Gal3 Abs in mice induced local inflammation with perivascular infiltration of T cells and neutrophils, which was inhibited by IL-1β blockade. Induction of anti-Gal3 Abs in circulation by immunization of Gal3 antigen not only led to histopathologic changes in the skin, including focal keratinocytes vacuolization and thickening of blood vessels, but also a systemic autoimmune phenotype that involves autoantibody production and kidney damage. Intriguingly, local overexpression of IL-1β was primarily associated with skin lesions but not with other internal organs in mice. Finally, we showed that the serum levels of IL-1β were comparable between SLE patients and healthy donors. Whilst the expression of IL-1β was enriched in local area with perivascular inflammation in lupus skin lesion compared to healthy normal skin. The results strongly suggest that IL-1β plays an important role in mediating anti-Gal3 Ab-induced skin vascular inflammation and raised the prospect for using IL-1β blocking therapies to treat lupus cutaneous damage.
Collapse
Affiliation(s)
- Zhenrui Shi
- Department of Dermatology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Yu-Ping Zhang
- Department of Dermatology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China; Department of Dermatology, Zhongshan People's Hospital, Zhongshan, Guangdong, China
| | - Dan Hong
- Department of Dermatology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Xiaonan Qiu
- Department of Dermatology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Lin Zheng
- Department of Dermatology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China; Department of Dermatology, Chinese Academy of Medical Sciences & Peking Union Medical College Hospital of Skin Diseases and Institute of Dermatology, Nanjing, Jiangsu, China
| | - Lijuan Bian
- Department of Pathology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Fengqiu Hu
- Department of Dermatology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Liuyu Chen
- Department of Dermatology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China; Department of Dermatology, Sun Yat-sen University 8th Affiliated Hospital, Shenzhen, China
| | - Hui Xiong
- Department of Dermatology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Qiongqiong Yang
- Department of Nephrology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Shanping Jiang
- Department of Respiration, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Guozhen Tan
- Department of Dermatology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Liangchun Wang
- Department of Dermatology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China.
| |
Collapse
|
30
|
Gharamti AA, Samara O, Monzon A, Montalbano G, Scherger S, DeSanto K, Chastain DB, Sillau S, Montoya JG, Franco-Paredes C, Henao-Martínez AF, Shapiro L. Proinflammatory cytokines levels in sepsis and healthy volunteers, and tumor necrosis factor-alpha associated sepsis mortality: A systematic review and meta-analysis. Cytokine 2022; 158:156006. [PMID: 36044827 DOI: 10.1016/j.cyto.2022.156006] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Revised: 07/09/2022] [Accepted: 08/05/2022] [Indexed: 01/10/2023]
Abstract
BACKGROUND Sepsis is a global health challenge associated with significant morbidity and mortality. Detrimental sepsis effects are attributed to excessive inflammation or a "cytokine storm." However, anti-inflammation therapies have failed to lower sepsis mortality. We aim to characterize levels of key inflammatory cytokines in patients with sepsis and compare levels with those in healthy individuals and relate tumor necrosis factor (TNF) α levels to patient characteristics and outcomes. METHODS We performed a systematic review and meta-analysis. Medline, Embase, Cochrane Library, and Web of Science Core Collection databases were searched between 1985 and May 2020. Analysis was restricted to studies in English. We included randomized controlled trials (RCTs), controlled trials, cohort studies, case series, and cross-sectional studies that reported mean levels of cytokines in the circulation thought to be relevant for sepsis pathogenesis. We also evaluated concentrations of these cytokines in healthy individuals. The Quality in Prognosis Studies tool was used to assess the methodological quality of included studies. We extracted summary data from published reports. Data analyses were performed using a random-effects model to estimate pooled odds ratios (OR) with 95% confidence intervals for cytokine levels and mortality. This systematic review is registered in PROSPERO (CRD42020179800). FINDINGS We identified 3654 records, and 104 studies were included with a total of 3250 participants. The pooled estimated mean TNFα concentration in sepsis patients was 58.4 pg/ml (95% Confidence Interval or CI 39.8-85.8 pg/ml), and in healthy individuals was 5.5 pg/ml (95% CI 3.8-8.0 pg/ml). Pooled estimate means for IL-1β and IFN-γ in sepsis patients were 21.8 pg/ml and 63.3 pg/ml, respectively. Elevated TNFα concentrations associated with increased 28-day sepsis mortality (p = 0.001). In subgroup analyses, we did not detect an association between TNFα levels and sepsis source, sepsis severity, or sequential organ failure assessment (SOFA) score. A TNF-α cutoff level ≥14.7 pg/ml separated sepsis patients from healthy individuals with a sensitivity of 82.6%, a specificity of 91.7%, and a likelihood ratio of 9.9. INTERPRETATION Sepsis mean TNFα concentration is increased approximately 10-fold compared to mean concentration in healthy individuals, and TNFα associated with sepsis mortality but not sepsis severity. The concept that elevated cytokines cause sepsis should be revisited in the context of these data. FUNDING None.
Collapse
Affiliation(s)
- Amal A Gharamti
- Department of Internal Medicine, Yale University, Waterbury Hospital, Waterbury, CT, USA
| | - Omar Samara
- School of Medicine, Division of Infectious Diseases, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Anthony Monzon
- School of Medicine, Division of Infectious Diseases, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Gabrielle Montalbano
- School of Medicine, Division of Infectious Diseases, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Sias Scherger
- School of Medicine, Division of Infectious Diseases, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Kristen DeSanto
- Strauss Health Sciences Library, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Daniel B Chastain
- Department of Clinical and Administrative Pharmacy, University of Georgia College of Pharmacy, Albany, GA, USA
| | - Stefan Sillau
- Department of Neurology and Department of Biostatistics, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Jose G Montoya
- Dr. Jack S. Remington Laboratory for Specialty Diagnostics, Palo Alto, CA, USA
| | - Carlos Franco-Paredes
- School of Medicine, Division of Infectious Diseases, University of Colorado Anschutz Medical Campus, Aurora, CO, USA; Hospital Infantil de México, Federico Gómez, Mexico City, Mexico
| | - Andrés F Henao-Martínez
- School of Medicine, Division of Infectious Diseases, University of Colorado Anschutz Medical Campus, Aurora, CO, USA.
| | - Leland Shapiro
- School of Medicine, Division of Infectious Diseases, University of Colorado Anschutz Medical Campus, Aurora, CO, USA; Rocky Mountain Regional Veterans Affairs Medical Center, Aurora, CO, USA
| |
Collapse
|
31
|
Gao Q, Wang Y, Li J, Bai G, Liu L, Zhong R, Ma T, Pan H, Zhang H. Supplementation of multi-enzymes alone or combined with inactivated Lactobacillus benefits growth performance and gut microbiota in broilers fed wheat diets. Front Microbiol 2022; 13:927932. [PMID: 35979486 PMCID: PMC9376439 DOI: 10.3389/fmicb.2022.927932] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Accepted: 06/30/2022] [Indexed: 11/13/2022] Open
Abstract
The effects of multi-enzymes mixture supplementation or combination with inactivated Lactobacillus on growth performance, intestinal barrier, and cecal microbiota were investigated in broilers at the age of 15-42 days fed a wheat-based diet. A total of 576 broilers (12 broilers/cage; n = 12) were used and divided into four groups and randomly allotted to four experimental diets throughout grower (15-28 days of age) and finisher (29-42 days of age) phases. Diets consisted of a corn-soybean meal-based diet (BD), a wheat-soybean meal-based diet (WD), and WD supplemented multi-enzymes (WED) or combined with inactivated Lactobacillus (WEPD). The results showed that the average daily gain (ADG) and body weight (BW) were reduced in broilers fed WD diet compared with those fed BD diet during the grower period (P < 0.05). Broilers in the WED or WEPD group had higher ADG and BW during the grower period (P < 0.05) and had a lower feed-to-gain ratio (F/G) compared to broilers in the WD group during the grower and overall periods (P < 0.05). Improved expression of intestinal barrier genes (claudin-1, ZO-1, and mucin-2) was observed in WEPD compared to the BD or WD group (P < 0.05). Compared to the BD group, the WD group decreased the abundance of Oscillospira, norank_f__Erysipelotrichaceae, and Peptococcus, which are related to anti-inflammatory function and BW gain. The WD also increased Bifidobacterium and some short-chain fatty acid (SCFA)-producing bacteria (Anaerotruncus, Blautia, and Oscillibacter), and Barnesiella, which were presumed as "harmful microbes" [false discovery rate (FDR) < 0.05]. WED and WEPD groups, respectively, improved Bilophila and Eubacterium_hallii_group compared with those in the WD group (FDR < 0.05). In addition, the Enterococcus abundance was reduced in the WEPD group compared to the WD group (FDR < 0.05). Higher acetate and total SCFA concentrations were observed (P < 0.05) among broilers who received a WD diet. Compared with the WD group, the WED or WEPD group further increased cecal propionate content (P < 0.05) and tended to improve butyrate concentration. These results suggested that supplemental multi-enzymes alone and combined with inactivated Lactobacillus could improve the growth performance based on the wheat-based diet and offer additional protective effects on the intestinal barrier function of broilers.
Collapse
Affiliation(s)
- Qingtao Gao
- State Key Laboratory of Animal Nutrition, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Yanchun Wang
- Yunnan Provincial Key Laboratory of Animal Nutrition and Feed Science, College of Animal Science and Technology, Yunnan Agricultural University, Kunming, China
| | - Jiaheng Li
- State Key Laboratory of Animal Nutrition, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing, China
- Precision Livestock and Nutrition Unit, Gembloux Agro-Bio Tech, Liège University, Gembloux, Belgium
| | - Guosong Bai
- State Key Laboratory of Animal Nutrition, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Lei Liu
- State Key Laboratory of Animal Nutrition, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Ruqing Zhong
- State Key Laboratory of Animal Nutrition, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Teng Ma
- State Key Laboratory of Animal Nutrition, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Hongbin Pan
- Yunnan Provincial Key Laboratory of Animal Nutrition and Feed Science, College of Animal Science and Technology, Yunnan Agricultural University, Kunming, China
| | - Hongfu Zhang
- State Key Laboratory of Animal Nutrition, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing, China
| |
Collapse
|
32
|
Charoensappakit A, Sae-Khow K, Leelahavanichkul A. Gut Barrier Damage and Gut Translocation of Pathogen Molecules in Lupus, an Impact of Innate Immunity (Macrophages and Neutrophils) in Autoimmune Disease. Int J Mol Sci 2022; 23:ijms23158223. [PMID: 35897790 PMCID: PMC9367802 DOI: 10.3390/ijms23158223] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Revised: 07/25/2022] [Accepted: 07/25/2022] [Indexed: 02/08/2023] Open
Abstract
The gut barrier is a single cell layer that separates gut micro-organisms from the host, and gut permeability defects result in the translocation of microbial molecules from the gut into the blood. Despite the silent clinical manifestation, gut translocation of microbial molecules can induce systemic inflammation that might be an endogenous exacerbating factor of systemic lupus erythematosus. In contrast, circulatory immune-complex deposition and the effect of medications on the gut, an organ with an extremely large surface area, of patients with active lupus might cause gut translocation of microbial molecules, which worsens lupus severity. Likewise, the imbalance of gut microbiota may initiate lupus and/or interfere with gut integrity which results in microbial translocation and lupus exacerbation. Moreover, immune hyper-responsiveness of innate immune cells (macrophages and neutrophils) is demonstrated in a lupus model from the loss of inhibitory Fc gamma receptor IIb (FcgRIIb), which induces prominent responses through the cross-link between activating-FcgRs and innate immune receptors. The immune hyper-responsiveness can cause cell death, especially apoptosis and neutrophil extracellular traps (NETosis), which possibly exacerbates lupus, partly through the enhanced exposure of the self-antigens. Leaky gut monitoring and treatments (such as probiotics) might be beneficial in lupus. Here, we discuss the current information on leaky gut in lupus.
Collapse
Affiliation(s)
- Awirut Charoensappakit
- Center of Excellence in Translational Research in Inflammation and Immunology (CETRII), Department of Microbiology, Faculty of Medicine, Chulalongkorn University, Bangkok 10330, Thailand
| | - Kritsanawan Sae-Khow
- Center of Excellence in Translational Research in Inflammation and Immunology (CETRII), Department of Microbiology, Faculty of Medicine, Chulalongkorn University, Bangkok 10330, Thailand
| | - Asada Leelahavanichkul
- Center of Excellence in Translational Research in Inflammation and Immunology (CETRII), Department of Microbiology, Faculty of Medicine, Chulalongkorn University, Bangkok 10330, Thailand
- Nephrology Unit, Department of Medicine, Faculty of Medicine, Chulalongkorn University, Bangkok 10330, Thailand
| |
Collapse
|
33
|
Effect of add-on hydroxychloroquine therapy on serum proinflammatory cytokine levels in patients with systemic lupus erythematosus. Sci Rep 2022; 12:10175. [PMID: 35715525 PMCID: PMC9205904 DOI: 10.1038/s41598-022-14571-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Accepted: 06/08/2022] [Indexed: 11/17/2022] Open
Abstract
We investigated the effect of hydroxychloroquine (HCQ) as an add-on treatment to immunosuppressants on the expression of proinflammatory cytokines in patients with systemic lupus erythematosus. Serum levels of tumor necrosis factor (TNF)-α, interleukin (IL)-2, IL-6, IL-8, vascular endothelial growth factor (VEGF)-A, monocyte chemotactic protein-1 (MCP-1), macrophage inflammatory protein-1α (MIP-1α), and interleukin 1 receptor antagonist (IL-1ra) were measured immediately before and 3 months after treatment with oral HCQ. Among the 51 patients enrolled in the study, HCQ treatment led to significantly reduced serum levels of TNF-α, IL-6, IL-8, VEGF-A, IL-1ra, and IL-2 (p < 0.0001; p = 0.0006; p = 0.0460, p = 0.0177; p < 0.0001; p = 0.0282, respectively) and to decreased (but not significantly) levels of MIP-1α (p = 0.0746). No significant changes were observed in the serum MCP-1 levels before and after HCQ administration (p = 0.1402). Our results suggest that an add-on HCQ treatment modulates the expression of proinflammatory cytokines even in systemic lupus erythematosus patients with low disease activity.
Collapse
|
34
|
Longitudinal comparison of IL-6, IL-10, and IL-12 cytokine profiles in adult and childhood-onset systemic lupus erythematosus. J Transl Autoimmun 2022; 5:100158. [PMID: 36683864 PMCID: PMC9846463 DOI: 10.1016/j.jtauto.2022.100158] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Accepted: 05/20/2022] [Indexed: 01/25/2023] Open
Abstract
Objective To compare the levels of Th1 (IL-12) and Th2 (IL-6 and IL10) cytokines over a two-year period among systemic lupus erythematosus patients with childhood-onset (cSLE), adult-onset (sSLE), and healthy controls, and correlate with their clinical, laboratory, and treatment manifestations. Methods The study included 63 patients with cSLE [57 (90%) women; mean age 19.7 ± 4.3 years (range = 10-29); mean disease duration 7.3 ± 4.2 years (range 2-15)], 67 patients with aSLE [65 (97%) women; mean age of 39.9 ± 11.8 years (range 21-68); disease duration 7.7 ± 3.1 years (range 4-16)], and 40 healthy controls [36 (90%) women; mean age of 29.6 ± 10 years (range 12-49)]. cSLE and aSLE patients were paired by disease duration. Clinical and laboratory manifestations, disease activity (SLEDAI), cumulative damage (SDI), and current drug exposures were evaluated. Symptoms of anxiety and depression were evaluated by the Beck inventory (BAI and BDI, respectively). Th1 (IL-12) and Th2 (IL-6 and IL-10) cytokines were measured by the ELISA test. Data were collected at four different time points (TI, TII, TIII, and TIV) and compared by non-parametric tests. Results IL-6 levels were significantly higher in aSLE patients compared to healthy controls at times I, II, and III (TI p = 0.013, TII p = 0.015, TIII p = 0.004, and TIV p = 0.634). However, no difference was observed between cSLE patients and healthy controls (TI p = 0.223, TII p = 0.613, TIII p = 0.341, and TIV p = 0.977). In addition, no difference was observed between aSLE and cSLE patients (TI p = 0.377, TII p = 0.123, TIII p = 0.105, and TIV p = 0.591). The levels of IL-12 were significantly higher in cSLE patients compared to healthy controls at all time points (TI p = 0.04, TII p < 0.001, TIII p = 0.015, and TIV p = 0.021). aSLE patients showed significantly elevated levels when compared to healthy controls at time III and IV (TI p = 0.752, TII p = 0.827, TIII p = 0.011*, and TIV p < 0.001*). cSLE patients showed significantly higher levels than aSLE patients at times I and II (TI p = 0.07*, TII p < 0.001*, TIII p = 0.998, and TIV p = 0.140). In aSLE patients, IL-6 was associated with headache (p = 0.006), arthritis (p = 0.044), and nephritis (p = 0.012); IL-10 was associated with nephritis (p = 0.043), hypocomplementemia (p = 0.001), and disease activity (p = 0.001); in these patients, IL-12 was associated with alopecia (p = 0.025) and leukopenia (p = 0.044). In cSLE patients, IL-6 was associated with arthritis (p = 0.022) and malar rash (p = 0.012). Conclusion aSLE and cSLE patients with long disease duration present similar levels of cytokines, despite differences in clinical activity patterns over time.
Collapse
|
35
|
Moreno-Torres V, Castejón R, Martínez-Urbistondo M, Gutiérrez-Rojas Á, Vázquez-Comendador J, Tutor P, Durán-Del Campo P, Mellor-Pita S, Rosado S, Vargas-Núñez JA. Serum cytokines to predict systemic lupus erythematosus clinical and serological activity. Clin Transl Sci 2022; 15:1676-1686. [PMID: 35478259 PMCID: PMC9283738 DOI: 10.1111/cts.13283] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2022] [Revised: 03/22/2022] [Accepted: 03/31/2022] [Indexed: 12/22/2022] Open
Abstract
We aimed to explore the role of interleukin (IL)-6, interferon-gamma (IFNγ), IL-10, and tumor necrosis factor (TNF) as predictors of systemic lupus erythematosus (SLE) clinical and serological activity, and their correlation with the treatment received. We performed a retrospective analysis of 77 patients with SLE according to the 2012 Systemic Lupus International Collaborative Clinics (SLICC) criteria. The outcomes were serological activity (SA), active disease (AD), complete remission (CR), the low-disease activity state (LDAS), and immunosuppressive treatment. SA was present in 17.1%, AD in 17.3%, CR in 13%, and LDAS in 64.9% of patients. IL-6 values were higher in patients in SA, in AD, in those receiving steroids alone, and in patients without CR or LDAS (p < 0.05). IFNγ was associated with anti-double stranded DNA (dsDNA) antibodies positivity and immunosuppression, whereas IL-10 values were higher in patients with CR (p < 0.05). The IL6-IFN product was able to predict anti-double stranded DNA (anti-dsDNA) antibodies positivity (area under the receiver operating characteristic curve [AUC-ROC] = 0.705, 95% confidence interval [CI] 0.563-0.847), SA (AUC-ROC = 0.720, 95% CI 0.542-0.899), AD (AUC-ROC = 0.701, 95% CI 0.520-0.882), steroid treatment (AUC-ROC = 0.751, 95% CI 0.622-0.879), and the absence of LDAS (AUC-ROC = 0.700, 95% CI 0.558-0.834). The IL6-IFN/IL10 ratio predicted AD (AUC-ROC = 0.742, 955 CI 0.540-0.944), steroid treatment (AUC-ROC = 0.721, 95% CI 0.572-0.870), and the absence of LDAS (AUC-ROC = 0.694, 95% CI 0.536-0.853). In conclusion, IL-6, IL-10, and IFNγ might help to assess SLE serological and clinical activity. Their combination in the IL-6-IFN product and the IL-6xIFN to IL-10 ratio results in novel tools to determine and predict SA, AD, and LDAS. Prompt detection of SLE activity might allow a rapid intervention to avoid established or chronic damage.
Collapse
Affiliation(s)
- Victor Moreno-Torres
- Systemic Autoimmune Diseases Unit, Internal Medicine Service, IDIPHIM (University Hospital Puerta de Hierro Research Institute), Hospital Universitario Puerta de Hierro Majadahonda, Madrid, Spain
| | - Raquel Castejón
- Systemic Autoimmune Diseases Unit, Internal Medicine Service, IDIPHIM (University Hospital Puerta de Hierro Research Institute), Hospital Universitario Puerta de Hierro Majadahonda, Madrid, Spain
| | - María Martínez-Urbistondo
- Systemic Autoimmune Diseases Unit, Internal Medicine Service, IDIPHIM (University Hospital Puerta de Hierro Research Institute), Hospital Universitario Puerta de Hierro Majadahonda, Madrid, Spain
| | - Ángela Gutiérrez-Rojas
- Systemic Autoimmune Diseases Unit, Internal Medicine Service, IDIPHIM (University Hospital Puerta de Hierro Research Institute), Hospital Universitario Puerta de Hierro Majadahonda, Madrid, Spain
| | - Jose Vázquez-Comendador
- Systemic Autoimmune Diseases Unit, Internal Medicine Service, IDIPHIM (University Hospital Puerta de Hierro Research Institute), Hospital Universitario Puerta de Hierro Majadahonda, Madrid, Spain
| | - Pablo Tutor
- Systemic Autoimmune Diseases Unit, Internal Medicine Service, IDIPHIM (University Hospital Puerta de Hierro Research Institute), Hospital Universitario Puerta de Hierro Majadahonda, Madrid, Spain
| | - Pedro Durán-Del Campo
- Systemic Autoimmune Diseases Unit, Internal Medicine Service, IDIPHIM (University Hospital Puerta de Hierro Research Institute), Hospital Universitario Puerta de Hierro Majadahonda, Madrid, Spain
| | - Susana Mellor-Pita
- Systemic Autoimmune Diseases Unit, Internal Medicine Service, IDIPHIM (University Hospital Puerta de Hierro Research Institute), Hospital Universitario Puerta de Hierro Majadahonda, Madrid, Spain
| | - Silvia Rosado
- Systemic Autoimmune Diseases Unit, Internal Medicine Service, IDIPHIM (University Hospital Puerta de Hierro Research Institute), Hospital Universitario Puerta de Hierro Majadahonda, Madrid, Spain
| | - Juan-Antonio Vargas-Núñez
- Systemic Autoimmune Diseases Unit, Internal Medicine Service, IDIPHIM (University Hospital Puerta de Hierro Research Institute), Hospital Universitario Puerta de Hierro Majadahonda, Madrid, Spain
| |
Collapse
|
36
|
Qian L, Yang F, Lin X, Jiang S, Zhang Y, Tang Y. Pyrroloquinoline quinone ameliorates liver injury in mice induced by cyclophosphamide. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2022; 29:30383-30393. [PMID: 34997497 DOI: 10.1007/s11356-021-17990-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Accepted: 12/03/2021] [Indexed: 06/14/2023]
Abstract
The current study aimed to investigate the potential ameliorative effects of pyrroloquinoline quinone (PQQ) on cyclophosphamide (CTX)-induced liver injury in mice. The liver injury model was established by injecting mice with CTX (80 mg/kg/day). Liver function indices, antioxidant enzyme activities, and inflammatory cytokines were evaluated. In addition, protein expression levels of the nuclear factor E2-related factor 2 (Nrf2) and nuclear factor kappa-B (NF-κB) pathways in the liver tissues were determined using western blot. The results indicated that PQQ decreased the serum alanine aminotransferase (ALT), aspartate aminotransferase (AST), and the malondialdehyde (MDA), interleukin (IL)-1β, IL-6, tumor necrosis factor-α (TNF-α) levels in the liver tissues. Moreover, PQQ enhanced the activities of oxidative stress markers to alleviate CTX induced oxidative stress. Furthermore, the expression levels of heme oxygenase-1 (HO-1), glutamate-cysteine ligase modifier subunit (GCLM), and NAD(P)H quinone oxidoreductase 1 (NQO1) were significantly increased, and the expression levels of NF-κB p50, NF-κB p65, and inhibitor of NF-κB kinase alpha (IKKα) were significantly decreased after PQQ administration, suggesting that PQQ alleviated CTX-induced liver injury via activating the Nrf2-mediated antioxidant response pathway, and inhibiting the NF-κB-mediated inflammation pathway. Therefore, PQQ can be potentially used as a dietary supplement or functional foods for alleviating the CTX-induced liver injury.
Collapse
Affiliation(s)
- Li Qian
- Zhejiang Provincial Engineering Technology Research Center of Marine Biomedical Products, School of Food and Pharmacy, Zhejiang Ocean University, Zhoushan, 316022, People's Republic of China
| | - Fei Yang
- Hangzhou Women's Hospital (Hangzhou Maternity and Child Health Care Hospital), Neonatal Intensive Care Unit, Hangzhou, 310008, People's Republic of China
| | - Xinhui Lin
- Zhejiang Provincial Engineering Technology Research Center of Marine Biomedical Products, School of Food and Pharmacy, Zhejiang Ocean University, Zhoushan, 316022, People's Republic of China
| | - Su Jiang
- ECA Healthcare Inc, Shanghai, 201101, People's Republic of China
| | - Yun Zhang
- Qianjiang College, Hangzhou Normal University, Hangzhou, 310012, People's Republic of China.
| | - Yunping Tang
- Zhejiang Provincial Engineering Technology Research Center of Marine Biomedical Products, School of Food and Pharmacy, Zhejiang Ocean University, Zhoushan, 316022, People's Republic of China.
| |
Collapse
|
37
|
Bolouri N, Akhtari M, Farhadi E, Mansouri R, Faezi ST, Jamshidi A, Mahmoudi M. Role of the innate and adaptive immune responses in the pathogenesis of systemic lupus erythematosus. Inflamm Res 2022; 71:537-554. [PMID: 35298669 DOI: 10.1007/s00011-022-01554-6] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2021] [Revised: 02/08/2022] [Accepted: 02/16/2022] [Indexed: 12/16/2022] Open
Abstract
OBJECTIVE Systemic lupus erythematosus (SLE), the most common form of lupus, is a multisystemic rheumatic disease with different clinical features that generally affect women of childbearing age. The common symptoms of SLE are very similar to other autoimmune and non-autoimmune disorders, thereby it is known as a thousand faces disease. In this article, we are going to discuss some of the most updated information about immune system-related factors, cells, and cytokines involved in SLE pathogenesis. METHODS Different electronic databases, especially PubMed/MEDLINE, Scopus, and Google Scholar, were searched to review and analyze relevant literature on the role of innate and adaptive immune cells and cytokines in the pathogenesis of SLE. A search for relevant literature was accomplished using various keywords including systemic lupus erythematosus, apoptosis, autoantibodies, immunopathogenesis of SLE, adaptive and innate immune cells, inflammatory cytokines, hormones, etc. RESULTS AND CONCLUSION: The most important characteristic of SLE is the production of antibodies against different nuclear autoantigens like double-strand DNA and RNA. The depositions of the immune complexes (ICs) that are generated between autoantibodies and autoantigens, along with aberrant clearance of them, can lead to permanent inflammation and contribute to tissue or organ damage. Related mechanisms underlying the initiation and development of SLE have not been clarified yet. Although, defects in immune tolerance, enhanced antigenic load, hyperactivity of T cells, and inappropriate regulation of B cells contribute to the pathogenic autoantibodies generation. Besides, sex hormones that influence the immune system seem to act as triggers or protectors of SLE development.
Collapse
Affiliation(s)
- Nasim Bolouri
- Immunology Department, Shahid Sadoughi University of Medical Sciences, Yazd, Iran.,Rheumatology Research Center, Shariati Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Maryam Akhtari
- Rheumatology Research Center, Shariati Hospital, Tehran University of Medical Sciences, Tehran, Iran.,Inflammation Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Elham Farhadi
- Rheumatology Research Center, Shariati Hospital, Tehran University of Medical Sciences, Tehran, Iran. .,Inflammation Research Center, Tehran University of Medical Sciences, Tehran, Iran.
| | - Reza Mansouri
- Immunology Department, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Seyedeh Tahereh Faezi
- Rheumatology Research Center, Shariati Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Ahmadreza Jamshidi
- Rheumatology Research Center, Shariati Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Mahdi Mahmoudi
- Rheumatology Research Center, Shariati Hospital, Tehran University of Medical Sciences, Tehran, Iran.,Inflammation Research Center, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
38
|
Zhang J, Chang L, Sun Y, Qin M, Wang X, Guo Y. Disabled-2 (DAB2) overexpression mediates immune suppression in systemic lupus erythematosus by modulating Treg/Th17 cell differentiation. Clin Exp Pharmacol Physiol 2022; 49:596-607. [PMID: 35108421 DOI: 10.1111/1440-1681.13630] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Revised: 12/27/2021] [Accepted: 01/25/2022] [Indexed: 10/19/2022]
Abstract
Systemic lupus erythematosus (SLE) is an autoimmune disorder. T helper 17 (Th17) and regulatory T (Treg) cells play key roles in SLE progression. Disabled-2 (DAB2) exhibits immunomodulatory effects in inflammatory diseases. However, the role of DAB2 in SLE and the precise mechanisms remain unknown. Herein, a decreased DAB2 expression and an increased miR-448-3p level were observed in PBMCs from SLE patients. DAB2 level was negatively correlated with SLE Disease Activity Index (SLEDAI), suggesting a functional correlation between DAB2 and SLE. To test this, we employed 8-week-old MRL/lpr mice and treated them with lentivirus-mediated DAB2 or its negative control (LV-NC). LV-DAB2 treatment increased DAB2 expression and reduced serum IgG and anti-dsDNA IgG levels. DAB2 up-regulation alleviated splenomegaly and lymphadenopathy and SLE-related organ damage. Moreover, DAB2 enhanced the percentage of CD25+ Foxp3+ Treg cells but reduced Th17 cell frequency in lupus, along with the reduction in TNF-α, IL-6 and IL-17A levels, and the elevation in IL-10. In vitro, naive CD4+ T cells isolated from MRL/lpr mice were polarized into Th17 or Treg phenotypes and treated with lentivirus. LV-DAB2 treatment down-regulated IL-17A expression and inhibited the generation of CD4+ IL-17A+ Th17 cells. Also, DAB2 triggered the production of IL-10 and the activation of Treg cells. Furthermore, DAB2 was verified as a direct target for miR-448-3p. MiR-448-3p overexpression canceled the promoting effect of DAB2 on Treg cell differentiation. Taken together, DAB2 exerts an immunosuppressive effect on SLE through promoting Treg cell activation and inhibiting Th17 cell differentiation, which may be modulated by miR-448-3p.
Collapse
Affiliation(s)
- Jing Zhang
- Department of Immunology and Rheumatology, Shengjing Hospital of China Medical University, Shenyang, 110022, Liaoning, People's Republic of China
| | - Lihua Chang
- Department of Immunology and Rheumatology, Shengjing Hospital of China Medical University, Shenyang, 110022, Liaoning, People's Republic of China
| | - Yue Sun
- Department of Immunology and Rheumatology, Shengjing Hospital of China Medical University, Shenyang, 110022, Liaoning, People's Republic of China
| | - Muting Qin
- Department of Immunology and Rheumatology, Shengjing Hospital of China Medical University, Shenyang, 110022, Liaoning, People's Republic of China
| | - Xiaofei Wang
- Department of Immunology and Rheumatology, Shengjing Hospital of China Medical University, Shenyang, 110022, Liaoning, People's Republic of China
| | - Yun Guo
- Department of Immunology and Rheumatology, Shengjing Hospital of China Medical University, Shenyang, 110022, Liaoning, People's Republic of China
| |
Collapse
|
39
|
Immunogenetics of Lupus Erythematosus. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2022; 1367:213-257. [DOI: 10.1007/978-3-030-92616-8_9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
|
40
|
Hashemi S, Habibagahi Z, Heidari M, Abdollahpour-Alitappeh M, Karimi MH. Effects of combined aerobic and anaerobic exercise training on cytokine profiles in patients with systemic lupus erythematosus (SLE); a randomized controlled trial. Transpl Immunol 2021; 70:101516. [PMID: 34922023 DOI: 10.1016/j.trim.2021.101516] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2021] [Revised: 12/11/2021] [Accepted: 12/12/2021] [Indexed: 11/19/2022]
Abstract
BACKGROUND Systemic lupus erythematosus (SLE) is an inflammatory rheumatic disease characterized by production of autoantibodies and organ damage. Elevated levels of cytokines have been reported in SLE patients. Physical activity could be considered one of the factors that affect the immune system status and function. The aim of the present study was to evaluate the effects of an 8-week supervised aerobic and anaerobic training program on the immune system of SLE patients through evaluation of serum cytokine levels. METHODS This cross-sectional study included 24 SLE patients selected between September 2015 and March 2016. The patients were randomly divided into two groups, including exercise (n = 14) and control (n = 10) groups. The exercise group participated in an 8-week combined supervised exercise training program consisting of three times per week in 60-min exercise sessions. After collection of whole peripheral blood, peripheral blood mononuclear cells (PBMCs) were isolated from the blood samples. Following RNA extraction and cDNA synthesis, the expression levels of IFN-γ, TNF-α, IL-6, IL-2, IL-4, IL-5, IL-9, IL-10, IL-13, IL-17A, IL-17F, IL-21, and IL-22 were determined using in-house SYBER Green-based real-time polymerase chain reaction (PCR). Lastly, the data obtained were analyzed using t-test. RESULTS The mean and standard deviation of age were 29.00 ± 3.19 and 21.50 ± 5.52 in the intervention and control groups, respectively. No significant differences were found among the mean serum levels of IFN-γ, IL6, IL-9, IL-17A, IL-17F and IL-21 among SLE patients in the intervention and control groups. The mean serum levels of TNF-α, IL2, IL-4, and IL-5 decreased significantly in the intervention as compared with the control group. The mean serum levels of IL-10, IL-13 and IL-22 significantly increased in the control group after eight weeks, as compared with the intervention group. CONCLUSIONS Our findings indicated that the 8-week supervised aerobic and anaerobic training program could result in decreased inflammatory cytokines.
Collapse
Affiliation(s)
- Somayeh Hashemi
- Department of Exercise Sciences, Shiraz University, Shiraz, Iran
| | - Zahra Habibagahi
- Department of Internal Medicine, Shiraz University of Medical Sciences, Iran
| | - Mojdeh Heidari
- Transplant Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | | | | |
Collapse
|
41
|
Raymond WD, Hamdorf M, Furfaro M, Eilertsen GO, Nossent JC. Smoking associates with increased BAFF and decreased interferon-γ levels in patients with systemic lupus erythematosus. Lupus Sci Med 2021; 8:8/1/e000537. [PMID: 34725185 PMCID: PMC8562512 DOI: 10.1136/lupus-2021-000537] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Accepted: 10/09/2021] [Indexed: 12/16/2022]
Abstract
Objective In SLE, smoking increases the burden of cutaneous disease and organ damage, and leads to premature mortality. However, the effect of smoking on disease manifestations and cytokine levels of patients with SLE is unclear. This study compared characteristics of patients with SLE across smoking status, and determined the association of smoking with serum cytokine levels. Method A cross-sectional study of patients with SLE (n=99) during a research visit in which smoking status was ascertained. Smoking status was compared across classification criteria (American College of Rheumatology Classification Criteria for SLE (ACR97)), disease activity (SLE Disease Activity Index), autoantibody levels, accrued damage (Systemic Lupus International Collaborating Clinics/ACR Damage Index), and circulating concentrations of serum interferon-gamma (IFN-γ), interleukin (IL)-1β, IL-4, IL-6, IL-10, IL-12, IL-17, B cell-activating factor (BAFF), tumour necrosis factor-alpha, transforming growth factor beta 1 (TGF-β1), macrophage inflammatory protein 1 alpha (MIP-1α), MIP-1β and monocyte chemoattractant protein 1. Linear regression models determined the association between smoking and cytokine levels, adjusting for age and sex, clinical characteristics (model 1), and anti-inflammatory (IL-4, IL-10 and TGF- β1) and regulatory (IL-1β) cytokines (model 2). Results Among patients with SLE (97.9% ANA+; mean 48.48 years old; 86.9% female; mean 10 years of disease duration), 35.4% (n=35 of 99) were smoking (an average of 7 cigarettes/day for 24 years). Smokers had increased odds of prevalent ACR97 malar rash (OR 3.40, 95% CI 1.23 to 9.34) and mucosal ulcers (OR 3.31, 95% CI 1.36 to 8.05). Smokers had more arthritis (OR 3.19, 95% CI 1.19 to 8.60), migraine (OR 2.82, 95% CI 1.07 to 7.44), Raynaud’s phenomenon (OR 5.15, 95% CI 1.95 to 13.56) and increased non-steroidal anti-inflammatory drug use (OR 6.88, 95% CI 1.99 to 23.72). Smoking associated with 27% increased BAFF levels (95% CI 6% to 48%) and 42% decreased IFN-γ levels (95% CI −79% to −5%) in model 2. Conclusion In patients with SLE, smoking independently associated with increased BAFF and decreased IFN-γ levels, and an increased frequency of arthritis, migraine and Raynaud’s phenomenon. Smoking cessation is advisable to reduce systemic inflammation, reduce disease activity and improve host defence.
Collapse
Affiliation(s)
- Warren David Raymond
- Rheumatology Section, Medical School, University of Western Australia Faculty of Medicine, Dentistry and Health Sciences, Crawley, Western Australia, Australia
| | - Matthew Hamdorf
- Rheumatology Section, Medical School, University of Western Australia Faculty of Medicine, Dentistry and Health Sciences, Crawley, Western Australia, Australia
| | - Michael Furfaro
- Rheumatology Section, Medical School, University of Western Australia Faculty of Medicine, Dentistry and Health Sciences, Crawley, Western Australia, Australia
| | | | - Johannes Cornelis Nossent
- Rheumatology Section, Medical School, University of Western Australia Faculty of Medicine, Dentistry and Health Sciences, Crawley, Western Australia, Australia.,Clinical Medicine, UiT The Arctic University of Norway, Tromso, Norway
| |
Collapse
|
42
|
Davies K, Dures E, Ng WF. Fatigue in inflammatory rheumatic diseases: current knowledge and areas for future research. Nat Rev Rheumatol 2021; 17:651-664. [PMID: 34599320 DOI: 10.1038/s41584-021-00692-1] [Citation(s) in RCA: 55] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/27/2021] [Indexed: 02/08/2023]
Abstract
Fatigue is a complex phenomenon and an important health concern for many people with chronic inflammatory rheumatic diseases, such as rheumatoid arthritis, psoriatic arthritis, primary Sjögren syndrome and systemic lupus erythematosus. Although some clinical trials have shown the benefits of cognitive behavioural therapy in fatigue management, the effect of this approach is relatively modest, and no curative treatment has been identified. The pathogenesis of fatigue remains unclear. Despite many challenges and limitations, a growing body of research points to roles for the immune system, the central and autonomic nervous systems and the neuroendocrine system in the induction and maintenance of fatigue in chronic diseases. New insights indicate that sleep, genetic susceptibility, metabolic disturbances and other biological and physiological mechanisms contribute to fatigue. Furthermore, understanding of the relationships between psychosocial factors and fatigue is increasing. However, the interrelationships between these diverse mechanisms and fatigue remain poorly defined. In this Review, we outline various biological, physiological and psychosocial determinants of fatigue in inflammatory rheumatic diseases, and propose mechanistic and conceptual models of fatigue to summarize current understanding, stimulate debate and develop further research ideas.
Collapse
Affiliation(s)
- Kristen Davies
- Translational and Clinical Research Institute, Newcastle University and NIHR Newcastle Biomedical Research Centre, Newcastle upon Tyne, UK.,Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle Upon Tyne, UK
| | - Emma Dures
- Academic Rheumatology, Bristol Royal Infirmary, Bristol, UK.,Faculty of Health and Applied Sciences, University of the West of England, Bristol, UK
| | - Wan-Fai Ng
- Translational and Clinical Research Institute, Newcastle University and NIHR Newcastle Biomedical Research Centre, Newcastle upon Tyne, UK. .,Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle Upon Tyne, UK.
| |
Collapse
|
43
|
Xiang N, Fang X, Sun XG, Zhou YB, Ma Y, Zhu C, Li XP, Wang GS, Tao JH, Li XM. Expression profile of PU.1 in CD4 +T cells from patients with systemic lupus erythematosus. Clin Exp Med 2021; 21:621-632. [PMID: 33966135 DOI: 10.1007/s10238-021-00717-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Accepted: 04/29/2021] [Indexed: 10/21/2022]
Abstract
Systemic lupus erythematosus (SLE) is a multisystem autoimmune disease with complex genetic predisposing factors involved. PU.1 is an important member of the ETS transcription factors family which has diverse functions such as regulating the proliferation, differentiation of immune cells and multiple inflammatory cytokines. Previous studies preliminary explored the relation between PU.1 and SLE. To further explain the potential role of PU.1 in the pathogenesis of SLE, 40 SLE patients and 20 age-sex matched healthy controls (HC) were recruited in this study. Flow cytometry was used to test the percentages of CD4+PU.1+T cells in peripheral blood mononuclear cells (PBMCs) from patients with SLE and HC. Expression levels of PU.1 mRNA in CD4+T cells from SLE patients and HC were analyzed by real-time transcription-polymerase chain reaction. Expression levels of plasma IL-1β, IL-9, IL-18, IL-6, IFN-α, TNF-α, IL-10 and TGF-β1 were measured by enzyme-linked immunosorbent assay. The percentage of CD4+PU.1+T cells in PBMCs from patients with SLE was significantly higher than that from HC (P < 0.001). In addition, the PU.1 mRNA expression in CD4+T cells from SLE patients was increased than that from HC (P = 0.002). In SLE patients, no significant correlation was found between the percentage of CD4+PU.1+T cells and the expression of PU.1 mRNA in CD4+T cells (P > 0.05). Associations of PU.1 mRNA expression in CD4+T cells with major clinical and laboratory parameters of SLE patients were also analyzed, but no significant correlations were found. Consistent with previous studies, SLE patients had increased IL-1β, IL-18, IL-6, IFN-α, TNF-α and IL-10 plasma concentrations than HC (P < 0.01). The expression level of plasma TGF-β1 was significantly decreased in SLE patients than in HC (P < 0.001). In SLE patients, the expression level of IL-1β was positive correlated with PU.1 mRNA expression in CD4+T cells (P = 0.001). Our study first time evaluated the expression profile of PU.1 in CD4+T cells from SLE patients confirming that PU.1 may participate in the pathogenesis of SLE.
Collapse
Affiliation(s)
- Nan Xiang
- Department of Rheumatology and Immunology, Anhui Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, 250012, Shandong, China
| | - Xuan Fang
- Department of Rheumatology and Immunology, Anhui Provincial Hospital, Hefei, 230001, Anhui, China
| | - Xiao-Ge Sun
- Department of Rheumatology and Immunology, Anhui Provincial Hospital, Hefei, 230001, Anhui, China
| | - Ying-Bo Zhou
- Department of Rheumatology and Immunology, Anhui Provincial Hospital, Hefei, 230001, Anhui, China
| | - Yan Ma
- Department of Rheumatology and Immunology, Anhui Provincial Hospital, Hefei, 230001, Anhui, China
| | - Chen Zhu
- Department of Rheumatology and Immunology, Anhui Provincial Hospital, Hefei, 230001, Anhui, China
| | - Xiang-Pei Li
- Department of Rheumatology and Immunology, Anhui Provincial Hospital, Hefei, 230001, Anhui, China
| | - Guo-Sheng Wang
- Department of Rheumatology and Immunology, Anhui Provincial Hospital, Hefei, 230001, Anhui, China
| | - Jin-Hui Tao
- Department of Rheumatology and Immunology, Anhui Provincial Hospital, Hefei, 230001, Anhui, China
| | - Xiao-Mei Li
- Department of Rheumatology and Immunology, Anhui Provincial Hospital, Hefei, 230001, Anhui, China.
| |
Collapse
|
44
|
Wu YR, Hsing CH, Chiu CJ, Huang HY, Hsu YH. Roles of IL-1 and IL-10 family cytokines in the progression of systemic lupus erythematosus: Friends or foes? IUBMB Life 2021; 74:143-156. [PMID: 34668305 DOI: 10.1002/iub.2568] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 09/03/2021] [Accepted: 09/28/2021] [Indexed: 12/20/2022]
Abstract
Systemic lupus erythematosus (SLE) is a chronic autoimmune disease of unknown etiology that can affect nearly every organ system in the body. Besides genetic and environmental factors, unbalanced pro-inflammatory and anti-inflammatory cytokines contribute to immune dysregulation, trigger an inflammatory response, and induce tissue and organ damage. Inflammatory responses in SLE can be promoted and/or maintained by the availability of cytokines that are overproduced systemically and/or in local tissues. Several key cytokines have been considered potential targets for the reduction of chronic inflammation in SLE. Recent studies indicated that dysregulated production of several cytokines, including those of the IL-1 family and IL-10 family, orchestrate immune activation and self-tolerance, play critical roles in the pathogenesis of SLE. Among IL-1 family cytokines, IL-1, IL-18, IL-33, IL-36, IL-37, and IL-38 had been the most thoroughly investigated in SLE. Additionally, IL-10 family cytokines, IL-10, IL-20, IL-22, IL-26, IL-28, and IL-29 are dysregulated in SLE. Therefore, a better understanding of the initiation and progression of SLE may provide suitable novel targets for therapeutic intervention. In this review, we discuss the involvement of inflammation in the pathogenesis of SLE, with a focus on IL-1 family and IL-10 family cytokines, and highlight pathophysiological approaches and therapeutic potential for treating SLE.
Collapse
Affiliation(s)
- Yi-Rou Wu
- Institute of Clinical Medicine, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Chung-Hsi Hsing
- Department of Anesthesiology, Chi Mei Medical Center, Tainan, Taiwan.,Department of Medical Research, Chi Mei Medical Center, Tainan, Taiwan
| | - Chiao-Juno Chiu
- Graduate Institute of Clinical Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Hsin-Yi Huang
- Institute of Clinical Medicine, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Yu-Hsiang Hsu
- Institute of Clinical Medicine, College of Medicine, National Cheng Kung University, Tainan, Taiwan.,Clinical Medicine Research Center, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| |
Collapse
|
45
|
Li A, Guo F, Pan Q, Chen S, Chen J, Liu HF, Pan Q. Mesenchymal Stem Cell Therapy: Hope for Patients With Systemic Lupus Erythematosus. Front Immunol 2021; 12:728190. [PMID: 34659214 PMCID: PMC8516390 DOI: 10.3389/fimmu.2021.728190] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Accepted: 09/14/2021] [Indexed: 12/26/2022] Open
Abstract
Systemic lupus erythematosus (SLE) is a chronic autoimmune disease. Although previous studies have demonstrated that SLE is related to the imbalance of cells in the immune system, including B cells, T cells, and dendritic cells, etc., the mechanisms underlying SLE pathogenesis remain unclear. Therefore, effective and low side-effect therapies for SLE are lacking. Recently, mesenchymal stem cell (MSC) therapy for autoimmune diseases, particularly SLE, has gained increasing attention. This therapy can improve the signs and symptoms of refractory SLE by promoting the proliferation of Th2 and Treg cells and inhibiting the activity of Th1, Th17, and B cells, etc. However, MSC therapy is also reported ineffective in some patients with SLE, which may be related to MSC- or patient-derived factors. Therefore, the therapeutic effects of MSCs should be further confirmed. This review summarizes the status of MSC therapy in refractory SLE treatment and potential reasons for the ineffectiveness of MSC therapy from three perspectives. We propose various MSC modification methods that may be beneficial in enhancing the immunosuppression of MSCs in SLE. However, their safety and protective effects in patients with SLE still need to be confirmed by further experimental and clinical evidence.
Collapse
Affiliation(s)
- Aifen Li
- Key Laboratory of Prevention and Management of Chronic Kidney Disease of Zhanjiang City, Institute of Nephrology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Fengbiao Guo
- Key Laboratory of Prevention and Management of Chronic Kidney Disease of Zhanjiang City, Institute of Nephrology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Quanren Pan
- Key Laboratory of Prevention and Management of Chronic Kidney Disease of Zhanjiang City, Institute of Nephrology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Shuxian Chen
- Key Laboratory of Prevention and Management of Chronic Kidney Disease of Zhanjiang City, Institute of Nephrology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Jiaxuan Chen
- Key Laboratory of Prevention and Management of Chronic Kidney Disease of Zhanjiang City, Institute of Nephrology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Hua-Feng Liu
- Key Laboratory of Prevention and Management of Chronic Kidney Disease of Zhanjiang City, Institute of Nephrology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Qingjun Pan
- Key Laboratory of Prevention and Management of Chronic Kidney Disease of Zhanjiang City, Institute of Nephrology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| |
Collapse
|
46
|
Wang J, Liu S, Ma J, Piao X. Changes in Growth Performance and Ileal Microbiota Composition by Xylanase Supplementation in Broilers Fed Wheat-Based Diets. Front Microbiol 2021; 12:706396. [PMID: 34335542 PMCID: PMC8319766 DOI: 10.3389/fmicb.2021.706396] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Accepted: 06/11/2021] [Indexed: 12/12/2022] Open
Abstract
Xylanase exerts key roles in improving growth performance and intestinal health of broilers fed wheat-based diets. However, knowledge is limited regarding effects of xylanase supplementation on ileal microbiota in broilers. A total of 128 one-day-old broilers (initial BW 48.03 ± 0.33 g) were selected to investigate effects of xylanase (AT-xynA) on growth performance, ileal morphology, microbiota composition, immune response, antioxidant capacity, and endocrine peptide levels in broilers. Broilers were randomly allotted into two dietary treatments (n = 8), namely, a wheat-soybean basal diet and a basal diet with 4,000 U/kg AT-xynA (XY). On days 7, 14, 21, and 42, broilers were weighted and ileal tissues were sampled. Ileal digesta samples were collected for analyzing microbiota composition on days 21 and 42. The results showed that AT-xynA could improve average daily weight gain and average daily feed intake, and there were interactions between diet and age of broilers (p < 0.05). On days 21 and 42, xylanase supplementation decreased ileal microbiota α-diversity, and the relative abundance of potentially pathogenic microbiota, such as phylum Proteobacteria, family Moraxellaceae and Staphylococcaceae, genus Staphylococcus, Pseudomonas, Streptococcus, and Enterococcus, increased the abundance of Lactobacillus (p < 0.05). Moreover, the reduction in acetate concentration and abundance of short-chain fatty acid-producing bacteria was also observed in broilers from XY group (p < 0.05). AT-xynA increased ileal villus height, glucagon-like peptide-1, and insulin-like growth factor-1 concentrations and decreased interleukin-1β, interleukin-6, tumor necrosis factor-α, and malondialdehyde content in broilers, and these positive effects on intestinal health were greater in young broilers. In conclusion, xylanase supplementation to wheat-based diets could improve ileal intestinal morphology and immune function, and alleviate excess fermentation of bacteria, which may be related to changes of intestinal microbiota. In addition, the positive effects of xylanase on intestinal health were more pronounced in young broilers, thus contributing to subsequent improvement in growth performance of broilers.
Collapse
Affiliation(s)
- Jian Wang
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Sujie Liu
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Jiayu Ma
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Xiangshu Piao
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University, Beijing, China
| |
Collapse
|
47
|
Sakunrangsit N, Metheepakornchai P, Kumpunya S, Greenblatt MB, Leelahavanichkul A, Pisitkun P, Lotinun S. Etanercept prevents TNF-α mediated mandibular bone loss in FcγRIIb-/- lupus model. PLoS One 2021; 16:e0250215. [PMID: 33861790 PMCID: PMC8051757 DOI: 10.1371/journal.pone.0250215] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Accepted: 04/01/2021] [Indexed: 12/30/2022] Open
Abstract
Patients with systemic lupus erythematosus are at increased risk for alveolar bone loss due to periodontitis possibly as a result of a pathogenic immune response to oral bacteria and inflammation. The aim of the present study was to investigate whether an anti-TNF-α antagonist could prevent mandibular bone loss in the FcγRIIb-/- mouse model of lupus. Mice lacking FcγRIIb had decreased cancellous and cortical bone volume at 6 months of age. Etanercept increased cancellous but not cortical bone volume in WT and increased both cancellous bone volume and cortical thickness in FcγRIIb-deficient mice. FcγRIIb deficiency decreased mRNA levels for osteoblast marker genes, Osx, Col1a1 and Alp without any change in osteoclast marker genes. Etanercept increased Osx, Alp, and Ocn in both WT and FcγRIIb-/- mice. Osteoclast marker genes including TNF-α, Trap and RANKL/OPG ratio was decreased in WT. Serum markers of proinflammatory cytokines, TNF-α, IFNγ, IL-6, and IL-17A, were increased in FcγRIIb-/- mice and etanercept antagonized these effects in FcγRIIb-/- mice. Etanercept increased serum PTH levels in the FcγRIIb-/- mouse model of lupus. Our results suggest that deletion of FcγRIIb induces osteopenia by increasing the level of proinflammatory cytokines. Etanercept is effective in preventing mandibular bone loss in FcγRIIb-/- mice, suggesting that anti-TNF-α therapy may be able to ameliorate mandibular bone loss in SLE patients with periodontitis.
Collapse
Affiliation(s)
- Nithidol Sakunrangsit
- Skeletal Disorders Research Unit, Department of Physiology, Faculty of Dentistry, Chulalongkorn University, Bangkok, Thailand
| | - Piyanuch Metheepakornchai
- Skeletal Disorders Research Unit, Department of Physiology, Faculty of Dentistry, Chulalongkorn University, Bangkok, Thailand
| | - Sarinya Kumpunya
- Interdisciplinary Program of Biomedical Sciences, Graduate School, Chulalongkorn University, Bangkok, Thailand
| | - Matthew Blake Greenblatt
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, Research Division, Hospital for Special Surgery, New York, NY, United States of America
| | - Asada Leelahavanichkul
- Division of Immunology, Department of Microbiology, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Prapaporn Pisitkun
- Division of Allergy, Immunology, and Rheumatology, Department of Medicine, Faculty of Medicine, Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| | - Sutada Lotinun
- Skeletal Disorders Research Unit, Department of Physiology, Faculty of Dentistry, Chulalongkorn University, Bangkok, Thailand
- * E-mail:
| |
Collapse
|
48
|
Kim J, Hope CM, Perkins GB, Stead SO, Scaffidi JC, Kette FD, Carroll RP, Barry SC, Coates PT. Rapamycin and abundant TCR stimulation are required for the generation of stable human induced regulatory T cells. Clin Transl Immunology 2020; 9:e1223. [PMID: 33425354 PMCID: PMC7780108 DOI: 10.1002/cti2.1223] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Revised: 07/07/2020] [Accepted: 11/12/2020] [Indexed: 12/18/2022] Open
Abstract
OBJECTIVES Regulatory T cells (Tregs) are a vital sub-population of CD4+ T cells with major roles in immune tolerance and homeostasis. Given such properties, the use of regulatory T cells for immunotherapies has been extensively investigated, with a focus on adoptive transfer of ex vivo expanded natural Tregs (nTregs). For immunotherapies, induced Tregs (iTregs), generated in vitro from naïve CD4+ T cells, provide an attractive alternative, given the ease of generating cell numbers required for clinical dosage. While the combination of TGF-β, ATRA and rapamycin has been shown to generate highly suppressive iTregs, the challenge for therapeutic iTreg generation has been their instability. Here, we investigate the impact of rapamycin concentrations and α-CD3/CD28 bead ratios on human iTreg stability. METHODS We assess iTregs generated with various concentrations of rapamycin and differing ratios of α-CD3/CD28 beads for their differentiation, stability, expression of Treg signature molecules and T helper effector cytokines, and Treg-specific demethylation region (TSDR) status. RESULTS iTregs generated in the presence of TGF-β, ATRA, rapamycin and a higher ratio of α-CD3/CD28 beads were highly suppressive and stable upon in vitro re-stimulation. These iTregs exhibited a similar expression profile of Treg signature molecules and T helper effector cytokines to nTregs, in the absence of TSDR demethylation. CONCLUSION This work establishes a method to generate human iTregs which maintain stable phenotype and function upon in vitro re-stimulation. Further validation in pre-clinical models will be needed to ensure its suitability for applications in adoptive transfer.
Collapse
Affiliation(s)
- Juewan Kim
- The Department of Molecular & Biomedical ScienceThe School of Biological SciencesThe Faculty of SciencesThe University of AdelaideAdelaideSAAustralia
| | - Christopher M Hope
- Department of GastroenterologyWomen’s and Children’s HospitalAdelaideSAAustralia
- Molecular Immunology GroupRobinson Research InstituteSchool of MedicineThe University of AdelaideAdelaideSAAustralia
| | - Griffith B Perkins
- The Department of Molecular & Biomedical ScienceThe School of Biological SciencesThe Faculty of SciencesThe University of AdelaideAdelaideSAAustralia
| | - Sebastian O Stead
- Discipline of MedicineSchool of MedicineThe University of AdelaideAdelaideSAAustralia
- College of Medicine and Public HealthDiscipline of MedicineFlinders UniversityBedford ParkSAAustralia
| | - Jacqueline C Scaffidi
- Discipline of MedicineSchool of MedicineThe University of AdelaideAdelaideSAAustralia
| | - Francis D Kette
- Discipline of MedicineSchool of MedicineThe University of AdelaideAdelaideSAAustralia
- College of Medicine and Public HealthDiscipline of MedicineFlinders UniversityBedford ParkSAAustralia
| | - Robert P Carroll
- Discipline of MedicineSchool of MedicineThe University of AdelaideAdelaideSAAustralia
- Central Northern Adelaide Renal and Transplantation Service (CNARTS)The Royal Adelaide HospitalAdelaideSAAustralia
- Division of Medical SciencesUniversity of South AustraliaAdelaideSAAustralia
| | - Simon C Barry
- Department of GastroenterologyWomen’s and Children’s HospitalAdelaideSAAustralia
- Molecular Immunology GroupRobinson Research InstituteSchool of MedicineThe University of AdelaideAdelaideSAAustralia
| | - Patrick Toby Coates
- Discipline of MedicineSchool of MedicineThe University of AdelaideAdelaideSAAustralia
- Central Northern Adelaide Renal and Transplantation Service (CNARTS)The Royal Adelaide HospitalAdelaideSAAustralia
| |
Collapse
|
49
|
The role of endocan and selected pro-inflammatory cytokines in systemic lupus erythematosus. Postepy Dermatol Alergol 2020; 37:898-903. [PMID: 33603606 PMCID: PMC7874865 DOI: 10.5114/ada.2019.90060] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2019] [Accepted: 04/09/2019] [Indexed: 01/04/2023] Open
Abstract
INTRODUCTION Systemic lupus erythematosus (SLE) is a multisystem inflammatory autoimmune disease with a wide spectrum of clinical manifestations. Cytokines such as interleukin-1 (IL-1) and tumour necrosis factor α (TNF-α) are involved in its pathogenesis. Endocan is a novel marker of endothelial dysfunction and is likely to be engaged in proinflammatory processes in SLE. AIM To determine whether endocan serum concentration in SLE patients vary from healthy controls. MATERIAL AND METHODS The study included 36 patients with SLE. SLEDAI-2K score was used to assess disease activity. The control group comprised 23 healthy volunteers. ELISA kits were used to assess serum concentrations of endocan, IL-1β, TNF-α, vascular endothelial growth factor (VEGF) and high-sensitivity C reactive protein (hs-CRP). RESULTS The serum concentration of endocan was significantly higher (p < 0.001) in the SLE group than in healthy individuals. A positive correlation was found between serum levels of endocan and IL-1β (r = 0.47, p < 0.05). Active SLE patients (SLEDAI-2K score above 6 points) with an elevated total cholesterol level (above 5.17 mmol/l) were found to have VEGF concentration higher than those with a normal cholesterol level (p < 0.03). No other relevant relationships were found between the serum concentration of endocan, other laboratory parameters, anthropometric features, activity and duration of SLE. CONCLUSIONS A higher serum level of endocan in SLE patients indicates its possible role in the pathogenesis of the disease and reflects endothelial dysfunction. Our findings indicate that endocan could serve as a potential marker of endothelial dysfunction in SLE.
Collapse
|
50
|
Sweeney M, Corden B, Cook SA. Targeting cardiac fibrosis in heart failure with preserved ejection fraction: mirage or miracle? EMBO Mol Med 2020; 12:e10865. [PMID: 32955172 PMCID: PMC7539225 DOI: 10.15252/emmm.201910865] [Citation(s) in RCA: 100] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2020] [Revised: 07/30/2020] [Accepted: 08/14/2020] [Indexed: 12/11/2022] Open
Abstract
Cardiac fibrosis is central to the pathology of heart failure, particularly heart failure with preserved ejection fraction (HFpEF). Irrespective of the underlying profibrotic condition (e.g. ageing, diabetes, hypertension), maladaptive cardiac fibrosis is defined by the transformation of resident fibroblasts to matrix-secreting myofibroblasts. Numerous profibrotic factors have been identified at the molecular level (e.g. TGFβ, IL11, AngII), which activate gene expression programs for myofibroblast activation. A number of existing HF therapies indirectly target fibrotic pathways; however, despite multiple clinical trials in HFpEF, a specific clinically effective antifibrotic therapy remains elusive. Therapeutic inhibition of TGFβ, the master-regulator of fibrosis, has unfortunately proven toxic and ineffective in clinical trials to date, and new approaches are needed. In this review, we discuss the pathophysiology and clinical implications of interstitial fibrosis in HFpEF. We provide an overview of trials targeting fibrosis in HFpEF to date and discuss the promise of potential new therapeutic approaches and targets in the context of underlying molecular mechanisms.
Collapse
Affiliation(s)
- Mark Sweeney
- MRC‐London Institute of Medical SciencesHammersmith Hospital CampusLondonUK
- Wellcome Trust 4i/NIHR Clinical Research FellowImperial CollegeLondonUK
| | - Ben Corden
- MRC‐London Institute of Medical SciencesHammersmith Hospital CampusLondonUK
- National Heart Research Institute SingaporeNational Heart Centre SingaporeSingaporeSingapore
- Cardiovascular and Metabolic Disorders ProgramDuke‐National University of Singapore Medical SchoolSingaporeSingapore
- National Heart and Lung InstituteImperial College LondonLondonUK
| | - Stuart A Cook
- MRC‐London Institute of Medical SciencesHammersmith Hospital CampusLondonUK
- National Heart Research Institute SingaporeNational Heart Centre SingaporeSingaporeSingapore
- Cardiovascular and Metabolic Disorders ProgramDuke‐National University of Singapore Medical SchoolSingaporeSingapore
- National Heart and Lung InstituteImperial College LondonLondonUK
| |
Collapse
|