1
|
Zarimeidani F, Rahmati R, Mostafavi M, Darvishi M, Khodadadi S, Mohammadi M, Shamlou F, Bakhtiyari S, Alipourfard I. Gut Microbiota and Autism Spectrum Disorder: A Neuroinflammatory Mediated Mechanism of Pathogenesis? Inflammation 2024:10.1007/s10753-024-02061-y. [PMID: 39093342 DOI: 10.1007/s10753-024-02061-y] [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: 01/31/2024] [Revised: 04/28/2024] [Accepted: 05/21/2024] [Indexed: 08/04/2024]
Abstract
Autism spectrum disorder (ASD) is a neurodevelopmental disorder characterized by impairments in social communication and behavior, frequently accompanied by restricted and repetitive patterns of interests or activities. The gut microbiota has been implicated in the etiology of ASD due to its impact on the bidirectional communication pathway known as the gut-brain axis. However, the precise involvement of the gut microbiota in the causation of ASD is unclear. This study critically examines recent evidence to rationalize a probable mechanism in which gut microbiota symbiosis can induce neuroinflammation through intermediator cytokines and metabolites. To develop ASD, loss of the integrity of the intestinal barrier, activation of microglia, and dysregulation of neurotransmitters are caused by neural inflammatory factors. It has emphasized the potential role of neuroinflammatory intermediates linked to gut microbiota alterations in individuals with ASD. Specifically, cytokines like brain-derived neurotrophic factor, calprotectin, eotaxin, and some metabolites and microRNAs have been considered etiological biomarkers. We have also overviewed how probiotic trials may be used as a therapeutic strategy in ASD to reestablish a healthy balance in the gut microbiota. Evidence indicates neuroinflammation induced by dysregulated gut microbiota in ASD, yet there is little clarity based on analysis of the circulating immune profile. It deems the repair of microbiota load would lower inflammatory chaos in the GI tract, correct neuroinflammatory mediators, and modulate the neurotransmitters to attenuate autism. The interaction between the gut and the brain, along with alterations in microbiota and neuroinflammatory biomarkers, serves as a foundational background for understanding the etiology, diagnosis, prognosis, and treatment of autism spectrum disorder.
Collapse
Affiliation(s)
- Fatemeh Zarimeidani
- Students Research Committee, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Rahem Rahmati
- Students Research Committee, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Mehrnaz Mostafavi
- Faculty of Allied Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammad Darvishi
- School of Aerospace and Subaquatic Medicine, Infectious Diseases & Tropical Medicine Research Center (IDTMC), AJA University of Medical Sciences, Tehran, Iran
| | - Sanaz Khodadadi
- Student Research Committee, Tehran Medical Sciences Branch, Islamic Azad University, Tehran, Iran
| | - Mahya Mohammadi
- Student Research Committee, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Farid Shamlou
- School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Salar Bakhtiyari
- Feinberg Cardiovascular and Renal Research Institute, North Western University, Chicago. Illinois, USA
| | - Iraj Alipourfard
- Institute of Physical Chemistry, Polish Academy of Sciences, Marcin Kasprzaka 44/52, 01-224, Warsaw, Poland.
| |
Collapse
|
2
|
Huang K, Chen S, Yu LJ, Wu ZM, Chen QJ, Wang XQ, Li FF, Liu JM, Wang YX, Mao LS, Shen WF, Zhang RY, Shen Y, Lu L, Dai Y, Ding FH. Serum secreted phosphoprotein 1 level is associated with plaque vulnerability in patients with coronary artery disease. Front Immunol 2024; 15:1285813. [PMID: 38426091 PMCID: PMC10902157 DOI: 10.3389/fimmu.2024.1285813] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Accepted: 02/01/2024] [Indexed: 03/02/2024] Open
Abstract
Background Vulnerable plaque was associated with recurrent cardiovascular events. This study was designed to explore predictive biomarkers of vulnerable plaque in patients with coronary artery disease. Methods To reveal the phenotype-associated cell type in the development of vulnerable plaque and to identify hub gene for pathological process, we combined single-cell RNA and bulk RNA sequencing datasets of human atherosclerotic plaques using Single-Cell Identification of Subpopulations with Bulk Sample Phenotype Correlation (Scissor) and Weighted gene co-expression network analysis (WGCNA). We also validated our results in an independent cohort of patients by using intravascular ultrasound during coronary angiography. Results Macrophages were found to be strongly correlated with plaque vulnerability while vascular smooth muscle cell (VSMC), fibrochondrocyte (FC) and intermediate cell state (ICS) clusters were negatively associated with unstable plaque. Weighted gene co-expression network analysis showed that Secreted Phosphoprotein 1 (SPP1) in the turquoise module was highly correlated with both the gene module and the clinical traits. In a total of 593 patients, serum levels of SPP1 were significantly higher in patients with vulnerable plaques than those with stable plaque (113.21 [73.65 - 147.70] ng/ml versus 71.08 [20.64 - 135.68] ng/ml; P < 0.001). Adjusted multivariate regression analysis revealed that serum SPP1 was an independent determinant of the presence of vulnerable plaque. Receiver operating characteristic curve analysis indicated that the area under the curve was 0.737 (95% CI 0.697 - 0.773; P < 0.001) for adding serum SPP1 in predicting of vulnerable plaques. Conclusion Elevated serum SPP1 levels confer an increased risk for plaque vulnerability in patients with coronary artery disease.
Collapse
Affiliation(s)
- Ke Huang
- Department of Vascular and Cardiology, Rui Jin Hospital Shanghai Jiaotong University School of Medicine, Shanghai, China
- Institute of Cardiovascular Diseases, Shanghai Jiaotong University, School of Medicine, Shanghai, China
| | - Shuai Chen
- Department of Vascular and Cardiology, Rui Jin Hospital Shanghai Jiaotong University School of Medicine, Shanghai, China
- Institute of Cardiovascular Diseases, Shanghai Jiaotong University, School of Medicine, Shanghai, China
| | - Lin-Jun Yu
- Department of Vascular and Cardiology, Rui Jin Hospital Shanghai Jiaotong University School of Medicine, Shanghai, China
- Shanghai Clinical Research Center for Interventional Medicine, Shanghai, China
| | - Zhi-Ming Wu
- Department of Vascular and Cardiology, Rui Jin Hospital Shanghai Jiaotong University School of Medicine, Shanghai, China
- Institute of Cardiovascular Diseases, Shanghai Jiaotong University, School of Medicine, Shanghai, China
| | - Qiu-Jing Chen
- Institute of Cardiovascular Diseases, Shanghai Jiaotong University, School of Medicine, Shanghai, China
| | - Xiao-Qun Wang
- Department of Vascular and Cardiology, Rui Jin Hospital Shanghai Jiaotong University School of Medicine, Shanghai, China
- Institute of Cardiovascular Diseases, Shanghai Jiaotong University, School of Medicine, Shanghai, China
| | - Fei-Fei Li
- Department of Vascular and Cardiology, Rui Jin Hospital Shanghai Jiaotong University School of Medicine, Shanghai, China
- Institute of Cardiovascular Diseases, Shanghai Jiaotong University, School of Medicine, Shanghai, China
| | - Jing-Meng Liu
- Department of Vascular and Cardiology, Rui Jin Hospital Shanghai Jiaotong University School of Medicine, Shanghai, China
- Institute of Cardiovascular Diseases, Shanghai Jiaotong University, School of Medicine, Shanghai, China
| | - Yi-Xuan Wang
- Department of Vascular and Cardiology, Rui Jin Hospital Shanghai Jiaotong University School of Medicine, Shanghai, China
- Institute of Cardiovascular Diseases, Shanghai Jiaotong University, School of Medicine, Shanghai, China
| | - Lin-Shuang Mao
- Department of Vascular and Cardiology, Rui Jin Hospital Shanghai Jiaotong University School of Medicine, Shanghai, China
- Institute of Cardiovascular Diseases, Shanghai Jiaotong University, School of Medicine, Shanghai, China
| | - Wei-Feng Shen
- Department of Vascular and Cardiology, Rui Jin Hospital Shanghai Jiaotong University School of Medicine, Shanghai, China
- Institute of Cardiovascular Diseases, Shanghai Jiaotong University, School of Medicine, Shanghai, China
| | - Rui-Yan Zhang
- Department of Vascular and Cardiology, Rui Jin Hospital Shanghai Jiaotong University School of Medicine, Shanghai, China
- Institute of Cardiovascular Diseases, Shanghai Jiaotong University, School of Medicine, Shanghai, China
| | - Ying Shen
- Institute of Cardiovascular Diseases, Shanghai Jiaotong University, School of Medicine, Shanghai, China
| | - Lin Lu
- Department of Vascular and Cardiology, Rui Jin Hospital Shanghai Jiaotong University School of Medicine, Shanghai, China
- Institute of Cardiovascular Diseases, Shanghai Jiaotong University, School of Medicine, Shanghai, China
| | - Yang Dai
- Department of Vascular and Cardiology, Rui Jin Hospital Shanghai Jiaotong University School of Medicine, Shanghai, China
- Institute of Cardiovascular Diseases, Shanghai Jiaotong University, School of Medicine, Shanghai, China
| | - Feng-Hua Ding
- Department of Vascular and Cardiology, Rui Jin Hospital Shanghai Jiaotong University School of Medicine, Shanghai, China
- Shanghai Clinical Research Center for Interventional Medicine, Shanghai, China
| |
Collapse
|
3
|
Tian X, Yang W, Jiang W, Zhang Z, Liu J, Tu H. Multi-Omics Profiling Identifies Microglial Annexin A2 as a Key Mediator of NF-κB Pro-inflammatory Signaling in Ischemic Reperfusion Injury. Mol Cell Proteomics 2024; 23:100723. [PMID: 38253182 PMCID: PMC10879806 DOI: 10.1016/j.mcpro.2024.100723] [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: 06/01/2023] [Revised: 12/19/2023] [Accepted: 12/23/2023] [Indexed: 01/24/2024] Open
Abstract
Cerebral stroke is one of the leading causes of mortality and disability worldwide. Restoring the cerebral circulation following a period of occlusion and subsequent tissue oxygenation leads to reperfusion injury. Cerebral ischemic reperfusion (I/R) injury triggers immune and inflammatory responses, apoptosis, neuronal damage, and even death. However, the cellular function and molecular mechanisms underlying cerebral I/R-induced neuronal injury are incompletely understood. By integrating proteomic, phosphoproteomic, and transcriptomic profiling in mouse hippocampi after cerebral I/R, we revealed that the differentially expressed genes and proteins mainly fall into several immune inflammatory response-related pathways. We identified that Annexin 2 (Anxa2) was exclusively upregulated in microglial cells in response to cerebral I/R in vivo and oxygen-glucose deprivation and reoxygenation (OGD/R) in vitro. RNA-seq analysis revealed a critical role of Anxa2 in the expression of inflammation-related genes in microglia via the NF-κB signaling. Mechanistically, microglial Anxa2 is required for nuclear translocation of the p65 subunit of NF-κB and its transcriptional activity upon OGD/R in BV2 microglial cells. Anxa2 knockdown inhibited the OGD/R-induced microglia activation and markedly reduced the expression of pro-inflammatory factors, including TNF-α, IL-1β, and IL-6. Interestingly, conditional medium derived from Anxa2-depleted BV2 cell cultures with OGD/R treatment alleviated neuronal death in vitro. Altogether, our findings revealed that microglia Anxa2 plays a critical role in I/R injury by regulating NF-κB inflammatory responses in a non-cell-autonomous manner, which might be a potential target for the neuroprotection against cerebral I/R injury.
Collapse
Affiliation(s)
- Xibin Tian
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Biology, Hunan University, Changsha, Hunan, China
| | - Wuyan Yang
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Biology, Hunan University, Changsha, Hunan, China
| | - Wei Jiang
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Biology, Hunan University, Changsha, Hunan, China
| | - Zhen Zhang
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Biology, Hunan University, Changsha, Hunan, China
| | - Junqiang Liu
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Biology, Hunan University, Changsha, Hunan, China
| | - Haijun Tu
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Biology, Hunan University, Changsha, Hunan, China; Shenzhen Research Institute, Hunan University, Shenzhen, Guangdong, China.
| |
Collapse
|
4
|
Cicekli I, Saglam D, Takar N. A New Perspective on Metabolic Syndrome with Osteopontin: A Comprehensive Review. Life (Basel) 2023; 13:1608. [PMID: 37511983 PMCID: PMC10381599 DOI: 10.3390/life13071608] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Revised: 06/12/2023] [Accepted: 07/18/2023] [Indexed: 07/30/2023] Open
Abstract
Metabolic syndrome (MetS) imposes a substantial burden on the healthcare systems and economies of countries and is a major public health concern worldwide. MetS is mainly caused by an imbalance between calorie intake and energy expenditure; however, it is recognized that additional variables, such as chronic inflammation, may have the same predictive potential as insulin resistance or MetS components in the genesis of type 2 diabetes and cardiovascular events. More importantly, the early diagnosis or treatment of MetS may significantly reduce the burden on the health systems of the disease with any prevention or biomarker and should not be underestimated. Osteopontin (OPN), also called secreted phosphoprotein 1, is a soluble protein found mostly in body fluids. Studies suggest that serum OPN levels may be an early and new biomarker to predict metabolic and cardiovascular complications significantly associated with some diseases. This review aims to provide specific insight into the new biomarker OPN in MetS. With this purpose, it is examined the link between the MetS cornerstones and OPN. In addition, the interaction between the microbiota and MetS is predicted to be bidirectional, and the microbiota may act as a bridge in this interaction process. Increased OPN levels may have unfavourable consequences for cardiovascular diseases, diabetes, and obesity, all of which are components of MetS. Further studies are required to evaluate the use of OPN levels as a clinical biomarker risk of MetS.
Collapse
Affiliation(s)
- Ipek Cicekli
- Department of Nutrition and Dietetics, Faculty of Health Sciences, Acibadem Mehmet Ali Aydinlar University, Istanbul 34752, Turkey
| | - Duygu Saglam
- Department of Nutrition and Dietetics, Faculty of Health Sciences, Acibadem Mehmet Ali Aydinlar University, Istanbul 34752, Turkey
| | - Nadir Takar
- Department of Family Medicine, Kartal Dr. Lutfi Kirdar City Hospital, Istanbul Provincial Directorate of Health, Istanbul 34865, Turkey
| |
Collapse
|
5
|
Lin EYH, Xi W, Aggarwal N, Shinohara ML. Osteopontin (OPN)/SPP1: from its biochemistry to biological functions in the innate immune system and the central nervous system (CNS). Int Immunol 2023; 35:171-180. [PMID: 36525591 PMCID: PMC10071791 DOI: 10.1093/intimm/dxac060] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2022] [Accepted: 12/14/2022] [Indexed: 12/23/2022] Open
Abstract
Osteopontin (OPN) is a multifunctional protein, initially identified in osteosarcoma cells with its role of mediating osteoblast adhesion. Later studies revealed that OPN is associated with many inflammatory conditions caused by infections, allergic responses, autoimmunity and tissue damage. Many cell types in the peripheral immune system express OPN with various functions, which could be beneficial or detrimental. Also, more recent studies demonstrated that OPN is highly expressed in the central nervous system (CNS), particularly in microglia during CNS diseases and development. However, understanding of mechanisms underlying OPN's functions in the CNS is still limited. In this review, we focus on peripheral myeloid cells and CNS-resident cells to discuss the expression and functions of OPN.
Collapse
Affiliation(s)
- Elliot Yi-Hsin Lin
- Department of Immunology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Wen Xi
- Department of Immunology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Nupur Aggarwal
- Department of Immunology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Mari L Shinohara
- Department of Immunology, Duke University School of Medicine, Durham, NC 27710, USA
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, NC 27710, USA
| |
Collapse
|
6
|
Levy E, Marcil V, Tagharist Ép Baumel S, Dahan N, Delvin E, Spahis S. Lactoferrin, Osteopontin and Lactoferrin–Osteopontin Complex: A Critical Look on Their Role in Perinatal Period and Cardiometabolic Disorders. Nutrients 2023; 15:nu15061394. [PMID: 36986124 PMCID: PMC10052990 DOI: 10.3390/nu15061394] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Revised: 03/02/2023] [Accepted: 03/11/2023] [Indexed: 03/18/2023] Open
Abstract
Milk-derived bioactive proteins have increasingly gained attention and consideration throughout the world due to their high-quality amino acids and multiple health-promoting attributes. Apparently, being at the forefront of functional foods, these bioactive proteins are also suggested as potential alternatives for the management of various complex diseases. In this review, we will focus on lactoferrin (LF) and osteopontin (OPN), two multifunctional dairy proteins, as well as to their naturally occurring bioactive LF–OPN complex. While describing their wide variety of physiological, biochemical, and nutritional functionalities, we will emphasize their specific roles in the perinatal period. Afterwards, we will evaluate their ability to control oxidative stress, inflammation, gut mucosal barrier, and intestinal microbiota in link with cardiometabolic disorders (CMD) (obesity, insulin resistance, dyslipidemia, and hypertension) and associated complications (diabetes and atherosclerosis). This review will not only attempt to highlight the mechanisms of action, but it will critically discuss the potential therapeutic applications of the underlined bioactive proteins in CMD.
Collapse
Affiliation(s)
- Emile Levy
- Research Centre, CHU Sainte-Justine, 3175 Sainte-Catherine Road, Montreal, QC H3T 1C5, Canada
- Department of Nutrition, Université de Montreal, C. P. 6205, succursale Centre-ville, Montreal, QC H3C 3T5, Canada
| | - Valérie Marcil
- Research Centre, CHU Sainte-Justine, 3175 Sainte-Catherine Road, Montreal, QC H3T 1C5, Canada
- Department of Nutrition, Université de Montreal, C. P. 6205, succursale Centre-ville, Montreal, QC H3C 3T5, Canada
| | - Sarah Tagharist Ép Baumel
- Research Centre, CHU Sainte-Justine, 3175 Sainte-Catherine Road, Montreal, QC H3T 1C5, Canada
- Department of Nutrition, Université de Montreal, C. P. 6205, succursale Centre-ville, Montreal, QC H3C 3T5, Canada
| | - Noam Dahan
- Research Centre, CHU Sainte-Justine, 3175 Sainte-Catherine Road, Montreal, QC H3T 1C5, Canada
| | - Edgard Delvin
- Research Centre, CHU Sainte-Justine, 3175 Sainte-Catherine Road, Montreal, QC H3T 1C5, Canada
- Biochemistry &Molecular Medicine, Faculty of Medicine, Université de Montreal, C. P. 6205, succursale Centre-ville, Montreal, QC H3C 3T5, Canada
| | - Schohraya Spahis
- Research Centre, CHU Sainte-Justine, 3175 Sainte-Catherine Road, Montreal, QC H3T 1C5, Canada
- Biochemistry &Molecular Medicine, Faculty of Medicine, Université de Montreal, C. P. 6205, succursale Centre-ville, Montreal, QC H3C 3T5, Canada
- Correspondence: ; Tel.: +1-(514)-345-4832
| |
Collapse
|
7
|
Savran M, Ascı H, Erzurumlu Y, Ozmen O, Ilhan I, Sırın MC, Karakuyu NF, Karaibrahimoglu A. "Theranekron: A Novel Anti-inflammatory Candidate for Acetic Acid-Induced Colonic Inflammation in Rats". Mol Biol Rep 2022; 49:8753-8760. [PMID: 35939182 DOI: 10.1007/s11033-022-07722-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Revised: 06/13/2022] [Accepted: 06/15/2022] [Indexed: 11/26/2022]
Abstract
BACKGROUND Inflammatory bowel disease (IBD) is characterized with chronic inflammation of gastrointestinal track. In the pathogenesis of IBD, inflammation is the main mechanism. Induction of inflammation triggers the oxidative stress that subsequently leading to apoptosis. Considering the all pathological mechanisms, many therapeutic agents have been used for IBD but because of serious side effects there is still a need for new therapeutic drugs. In this study, we aim to evaluate the possible protective effects of Theranekron (TH) on acetic acid (AA)- induced colonic damage and to describe the probable effect mechanisms of TH. MATERIALS AND RESULTS Fourty female adult Wistar albino rats were divided into 5 groups. Following 24 h fasting, colitis was induced by rectal instillation of AA. In TH group, a single dose of subcutaneous 0.2 ml TH was used. In treatment groups, 0.2 ml TH single dose or 100 mg/kg sulfasalazine (SS) for 7 days were used after colitis induction. Normal salin was used for all applications in control group. Histopathologically hemorrhage, edema and inflammatory reactions were seen in AA group. TH and SS decreased the severity of lesions. Nuclear factor kappa B, Serum amyloid A, C-reactive protein, Growth-related oncogene, and Osteopontin expressions were markedly increased in AA group and TH markedly reduced these expressions. In Western analysis, decreased NF-kB and caspase-3 levels were observed with TH. Oxidative markers did not changed significantly. CONCLUSIONS TH has a prominent anti-inflammatory effect on AA-induced colonic inflammation via NF-kB signaling whereas antiapoptic effects seem to be independent from this pathway.
Collapse
Affiliation(s)
- Mehtap Savran
- Department of Pharmacology, Faculty of Medicine, Suleyman Demirel University, Isparta, Turkey.
| | - Halil Ascı
- Department of Pharmacology, Faculty of Medicine, Suleyman Demirel University, Isparta, Turkey
| | - Yalcin Erzurumlu
- Department of Biochemistry, Faculty of Pharmacy, Suleyman Demirel University, Isparta, Turkey
| | - Ozlem Ozmen
- Department of Pathology, Faculty of Veterinary Medicine, Mehmet Akif Ersoy University, Burdur, Turkey
| | - Ilter Ilhan
- Department of Medical Biochemistry, Faculty of Medicine, Suleyman Demirel University, Isparta, Turkey
| | - M Cem Sırın
- Department of Medical Microbiology, Faculty of Medicine, Suleyman Demirel University, Isparta, Turkey
| | - Nasif Fatih Karakuyu
- Department of Pharmacology, Faculty of Medicine, Suleyman Demirel University, Isparta, Turkey
| | - Adnan Karaibrahimoglu
- Department of Biostatistics and Medical Informatics, Faculty of Medicine, Süleyman Demirel University, Isparta, Turkey
| |
Collapse
|
8
|
Giannos P, Triantafyllidis KK, Giannos G, Kechagias KS. SPP1 in infliximab resistant ulcerative colitis and associated colorectal cancer: an analysis of differentially expressed genes. Eur J Gastroenterol Hepatol 2022; 34:598-606. [PMID: 35102110 DOI: 10.1097/meg.0000000000002349] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
OBJECTIVE Infliximab, a tumour necrosis factor-α (TNFα) antagonist, has advanced the management of ulcerative colitis. Although efficacious, considerable percentage of patients are resistant to treatment. Accumulative inflammatory burden in long-term ulcerative colitis patients refractory to therapy increases the risk of developing colorectal cancer (CRC). Our study investigated anti-TNFα-naïve patients with active ulcerative colitis to identify gene biomarkers whose dysregulated expression correlated with resistance to infliximab (IFX) treatment and poor prognosis in CRC. METHODS Differentially expressed genes (DEGs) from two studies (GSE73661 and GSE14580) with colonic mucosal samples were retrieved. Noninflammatory bowel disease controls were compared with those with active ulcerative colitis that either responded or were resistant to IFX before treatment. DEGs from ulcerative colitis samples resistant to IFX were used to construct a protein-protein interaction network, and clustering gene modules were identified. Module DEGs that overlapped with ulcerative colitis samples responsive to IFX were analysed, based on topological closeness and radiality. Hub genes were obtained, and their correlation with CRC progression was evaluated. Their expression in CRC tissues and their tumour microenvironment immune status was estimated. RESULTS Three clusters composed of 582 DEGs from ulcerative colitis samples resistant to IFX were retrieved. Comparative analysis identified 305 overlapping DEGs with ulcerative colitis samples responsive to IFX. Topological analysis revealed a hub gene - SPP1 - whose overexpression in CRC tissues and patients correlated with increased infiltration of immune signatures and poor prognosis. CONCLUSION SPP1 may serve as potential gene biomarker and predictor of resistance to IFX therapy in ulcerative colitis and CRC development.
Collapse
Affiliation(s)
- Panagiotis Giannos
- Department of Life Sciences, Faculty of Natural Sciences, Imperial College London
- Society of Meta-research and Biomedical Innovation, London
| | | | - Georgios Giannos
- Second Department of Surgery, Evaggelismos Hospital, Athens
- Department of Medicine, University of Crete Medical School, Heraklion, Crete, Greece
| | - Konstantinos S Kechagias
- Society of Meta-research and Biomedical Innovation, London
- Department of Metabolism, Digestion and Reproduction, Faculty of Medicine, Imperial College London
- Department of Obstetrics and Gynaecology, Chelsea and Westminster Hospital National Health Service (NHS) Foundation Trust, London, UK
| |
Collapse
|
9
|
Aggarwal N, Deerhake ME, DiPalma D, Shahi SK, Gaggioli MR, Mangalam AK, Shinohara ML. Secreted osteopontin from CD4 + T cells limits acute graft-versus-host disease. Cell Rep 2021; 37:110170. [PMID: 34965439 PMCID: PMC8759344 DOI: 10.1016/j.celrep.2021.110170] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2021] [Revised: 11/03/2021] [Accepted: 12/03/2021] [Indexed: 11/26/2022] Open
Abstract
Osteopontin (OPN) has been considered a potential biomarker of graft-versus-host disease (GVHD). However, the function of OPN in GVHD is still elusive. Using a mouse model of acute GVHD (aGVHD), we report that OPN generated by CD4+ T cells is sufficient to exert a beneficial effect in controlling aGVHD through limiting gastrointestinal pathology, a major target organ of aGVHD. CD4+ T cell-derived OPN works on CD44 expressed in intestinal epithelial cells (IECs) and abates cell death of IECs. OPN also modulates gut microbiota with enhanced health-associated commensal bacteria Akkermansia. Importantly, we use our in vivo mouse mutant model to specifically express OPN isoforms and demonstrate that secreted OPN (sOPN), not intracellular OPN (iOPN), is solely responsible for the protective role of OPN. This study demonstrates that sOPN generated by CD4+ T cells is potent enough to limit aGVHD.
Collapse
Affiliation(s)
- Nupur Aggarwal
- Department of Immunology, Duke University Medical School, Durham, NC 27710, USA
| | | | - Devon DiPalma
- Department of Immunology, Duke University Medical School, Durham, NC 27710, USA
| | - Shailesh K Shahi
- Department of Pathology, University of Iowa, Iowa City, IA 52242, USA
| | - Margaret R Gaggioli
- Department of Molecular Genetics and Microbiology, Duke University Medical School, Durham, NC 27710, USA
| | | | - Mari L Shinohara
- Department of Immunology, Duke University Medical School, Durham, NC 27710, USA; Department of Molecular Genetics and Microbiology, Duke University Medical School, Durham, NC 27710, USA.
| |
Collapse
|
10
|
Hansakon A, Png CW, Zhang Y, Angkasekwinai P. Macrophage-Derived Osteopontin Influences the Amplification of Cryptococcus neoformans-Promoting Type 2 Immune Response. THE JOURNAL OF IMMUNOLOGY 2021; 207:2107-2117. [PMID: 34526375 DOI: 10.4049/jimmunol.2100202] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/21/2021] [Accepted: 08/10/2021] [Indexed: 11/19/2022]
Abstract
A multifunctional glycoprotein, osteopontin (OPN), can modulate the function of macrophages, resulting in either protective or deleterious effects in various inflammatory diseases and infection in the lungs. Although macrophages play the critical roles in mediating host defenses against cryptococcosis or cryptococcal pathogenesis, the involvement of macrophage-derived OPN in pulmonary infection caused by fungus Cryptococcus has not been elucidated. Thus, our current study aimed to investigate the contribution of OPN to the regulation of host immune response and macrophage function using a mouse model of pulmonary cryptococcosis. We found that OPN was predominantly expressed in alveolar macrophages during C. neoformans infection. Systemic treatment of OPN during C. neoformans infection resulted in an enhanced pulmonary fungal load and an early onset of type 2 inflammation within the lung, as indicated by the increase of pulmonary eosinophil infiltration, type 2 cytokine production, and M2-associated gene expression. Moreover, CRISPR/Cas9-mediated OPN knockout murine macrophages had enhanced ability to clear the intracellular fungus and altered macrophage phenotype from pathogenic M2 to protective M1. Altogether, our data suggested that macrophage-derived OPN contributes to the elaboration of C. neoformans-induced type 2 immune responses and polarization of M2s that promote fungal survival and proliferation within macrophages.
Collapse
Affiliation(s)
- Adithap Hansakon
- Department of Medical Technology, Faculty of Allied Health Sciences, Thammasat University, Pathumthani, Thailand.,Graduate Program in Biomedical Science, Faculty of Allied Health Sciences, Thammasat University, Pathumthani, Thailand
| | - Chin Wen Png
- Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, Singapore, Singapore.,Immunology Programme, Life Science Institute, National University of Singapore, Singapore, Singapore; and
| | - Yongliang Zhang
- Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, Singapore, Singapore.,Immunology Programme, Life Science Institute, National University of Singapore, Singapore, Singapore; and
| | - Pornpimon Angkasekwinai
- Department of Medical Technology, Faculty of Allied Health Sciences, Thammasat University, Pathumthani, Thailand; .,Research Unit in Molecular Pathogenesis and Immunology of Infectious Diseases, Thammasat University, Pathumthani, Thailand
| |
Collapse
|
11
|
MyD88 in myofibroblasts enhances colitis-associated tumorigenesis via promoting macrophage M2 polarization. Cell Rep 2021; 34:108724. [PMID: 33535045 DOI: 10.1016/j.celrep.2021.108724] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2019] [Revised: 11/20/2020] [Accepted: 01/13/2021] [Indexed: 12/24/2022] Open
Abstract
The signal adaptor MyD88, an essential component of TLR signaling, plays an important role in gut-microbiome interactions. However, its contribution to colitis-associated cancer (CAC) is still controversial. Far less is known about the specific effects of MyD88 signaling in myofibroblasts in CAC development. Here, we used a CAC mouse model in which MyD88 was selectively depleted in myofibroblasts. Myofibroblast MyD88-deficient mice are resistant to azoxymethane (AOM)/dextran sodium sulfate (DSS)-induced tumorigenesis, as evidenced by the decrease in the number and sizes of tumors. MyD88 deficiency in myofibroblasts attenuates intestinal epithelial cell (IEC) proliferation after acute DSS-induced colitis. Furthermore, MyD88 signaling in myofibroblasts increases the secretion of osteopontin (OPN), which promotes macrophage M2 polarization through binding to αvβ3 and CD44, leading to activation of the STAT3/PPARγ pathway. Thus, MyD88 signaling in myofibroblasts crucially contributes to colorectal cancer development and provides a promising therapeutic target for the prevention of colitis-associated carcinogenesis.
Collapse
|
12
|
Smith BN, Hannas M, Orso C, Martins SMMK, Wang M, Donovan SM, Dilger RN. Dietary osteopontin-enriched algal protein as nutritional support in weaned pigs infected with F18-fimbriated enterotoxigenic Escherichia coli. J Anim Sci 2021; 98:5909275. [PMID: 32954424 DOI: 10.1093/jas/skaa314] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Accepted: 09/16/2020] [Indexed: 12/30/2022] Open
Abstract
This study investigated the effects of dietary osteopontin (OPN)-enriched algal protein on growth, immune status, and fecal fermentation profiles of weaned pigs challenged with a live infection of F18-fimbriated enterotoxigenic E. coli (ETEC). At 21 d of age, 54 pigs (5.95 ± 0.28 kg BW; blocked by BW) were allotted to 1 of 3 experimental groups combining dietary and health statuses. A control diet, containing 1% wild-type algal protein, was fed to both sham-inoculated (NC) and ETEC-inoculated (PC) pigs, while the test diet contained 1% OPN-enriched algal protein as fed only to ETEC-inoculated pigs (OA). All pigs received their assigned dietary treatment starting at study initiation to permit a 10-d acclimation period prior to inoculation. Growth performance, fecal dry matter, as well as hematological, histopathological, immune, and microbiota outcomes were analyzed by ANOVA, where treatment and time were considered as fixed effects and pig as a random effect; significance was accepted at P < 0.05. Overall, ETEC-inoculated pigs (PC and OA) exhibited decreased (P < 0.05) ADG and G:F, as well as increased (P < 0.05) peripheral blood helper T-cells and total leukocyte counts, compared with NC pigs during the postinoculation period. The OA treatment also elicited the highest (P < 0.05) concentrations of circulating tumor necrosis factor-α and volatile fatty acid concentrations in luminal contents at various postinoculation time-points, compared with other treatments. A principal coordinate analysis based on Unifrac weighted distances indicated that NC and OA groups had similar overall bacterial community structures, while PC pigs exhibited greater diversity, but infection status had no impact on α-diversity. Osteopontin-specific effects on microbial community structure included enrichment within Streptococcus and Blautia genera and decreased abundance of 12 other genera as compared with PC pigs. Overall, ETEC-infected pigs receiving 1% OPN-enriched algal protein exhibited changes immunity, inflammatory status, and colonic microbial community structure that may benefit weanling pigs experiencing F18 ETEC infection.
Collapse
Affiliation(s)
- Brooke N Smith
- Department of Animal Sciences, University of Illinois, Urbana, IL
| | - Melissa Hannas
- Department of Animal Science, Universidade Federal de Viçosa, Minas Gerais, Brazil
| | - Catiane Orso
- Department of Animal Sciences, University of Illinois, Urbana, IL
| | | | - Mei Wang
- Division of Nutritional Sciences, University of Illinois, Urbana, IL
| | - Sharon M Donovan
- Department of Food Science and Human Nutrition, University of Illinois, Urbana, IL.,Division of Nutritional Sciences, University of Illinois, Urbana, IL
| | - Ryan N Dilger
- Department of Animal Sciences, University of Illinois, Urbana, IL.,Department of Food Science and Human Nutrition, University of Illinois, Urbana, IL.,Neuroscience Program, University of Illinois, Urbana, IL
| |
Collapse
|
13
|
McQuitty CE, Williams R, Chokshi S, Urbani L. Immunomodulatory Role of the Extracellular Matrix Within the Liver Disease Microenvironment. Front Immunol 2020; 11:574276. [PMID: 33262757 PMCID: PMC7686550 DOI: 10.3389/fimmu.2020.574276] [Citation(s) in RCA: 68] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Accepted: 10/14/2020] [Indexed: 12/12/2022] Open
Abstract
Chronic liver disease when accompanied by underlying fibrosis, is characterized by an accumulation of extracellular matrix (ECM) proteins and chronic inflammation. Although traditionally considered as a passive and largely architectural structure, the ECM is now being recognized as a source of potent damage-associated molecular pattern (DAMP)s with immune-active peptides and domains. In parallel, the ECM anchors a range of cytokines, chemokines and growth factors, all of which are capable of modulating immune responses. A growing body of evidence shows that ECM proteins themselves are capable of modulating immunity either directly via ligation with immune cell receptors including integrins and TLRs, or indirectly through release of immunoactive molecules such as cytokines which are stored within the ECM structure. Notably, ECM deposition and remodeling during injury and fibrosis can result in release or formation of ECM-DAMPs within the tissue, which can promote local inflammatory immune response and chemotactic immune cell recruitment and inflammation. It is well described that the ECM and immune response are interlinked and mutually participate in driving fibrosis, although their precise interactions in the context of chronic liver disease are poorly understood. This review aims to describe the known pro-/anti-inflammatory and fibrogenic properties of ECM proteins and DAMPs, with particular reference to the immunomodulatory properties of the ECM in the context of chronic liver disease. Finally, we discuss the importance of developing novel biotechnological platforms based on decellularized ECM-scaffolds, which provide opportunities to directly explore liver ECM-immune cell interactions in greater detail.
Collapse
Affiliation(s)
- Claire E. McQuitty
- Institute of Hepatology, Foundation for Liver Research, London, United Kingdom
- Faculty of Life Sciences & Medicine, King’s College London, London, United Kingdom
| | - Roger Williams
- Institute of Hepatology, Foundation for Liver Research, London, United Kingdom
- Faculty of Life Sciences & Medicine, King’s College London, London, United Kingdom
| | - Shilpa Chokshi
- Institute of Hepatology, Foundation for Liver Research, London, United Kingdom
- Faculty of Life Sciences & Medicine, King’s College London, London, United Kingdom
| | - Luca Urbani
- Institute of Hepatology, Foundation for Liver Research, London, United Kingdom
- Faculty of Life Sciences & Medicine, King’s College London, London, United Kingdom
| |
Collapse
|
14
|
Wei F, Lang Y, Shen Q, Xu L, Cheng N, Chu Y, Lyu H, Chen F. Osteopontin-loaded PLGA nanoparticles enhance the intestinal mucosal barrier and alleviate inflammation via the NF-κB signaling pathway. Colloids Surf B Biointerfaces 2020; 190:110952. [PMID: 32172161 DOI: 10.1016/j.colsurfb.2020.110952] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2019] [Revised: 03/05/2020] [Accepted: 03/06/2020] [Indexed: 01/20/2023]
Abstract
Osteopontin is a multifunctional glycoprotein that is secreted by a variety of tissues or cells, but the role of osteopontin in the epithelial mucosal barrier has not been clearly established. We loaded osteopontin into hyaluronic acid-functionalized polymeric nanoparticles, which were administered by gavage to a colitis mouse model. The disease activity index, weight gain and colon length were calculated to assess the degree of symptoms. Epithelial permeability was measured using fluorescein isothiocyanate-conjugated dextran. The enzymatic activity of myeloperoxidase in the colon and inflammatory cytokines were assayed to assess the levels of inflammation. The histological appearance of the colon was observed by H&E staining. Tight junction proteins and signaling pathway proteins (NF-κB and phospho-NF-κB) were determined by western blotting. The resultant spherical osteopontin-loaded nanoparticles were characterized by the expected particle size (approximately 272.3 nm) and a slightly negative zeta potential (approximately -5.3 mV). Interestingly, we found that the osteopontin-loaded nanoparticles exerted remedial effects on colitis by both enhancing the intestinal barrier and alleviating inflammation in vivo according to the tested parameters. These results suggest that OPN plays a positive role in protecting the epithelial mucosal barrier and may be a therapeutic drug in gut homeostasis.
Collapse
Affiliation(s)
- Feng Wei
- Department of Gastroenterology, Shanghai Fifth People's Hospital, Fudan University, 128 Ruili Road, Shanghai 200240, People's Republic of China
| | - Yuhuang Lang
- Department of Emergency, Shanghai Fifth People's Hospital, Fudan University, 128 Ruili Road, Shanghai 200240, People's Republic of China
| | - Qiang Shen
- Department of Gastroenterology, Shanghai Fifth People's Hospital, Fudan University, 128 Ruili Road, Shanghai 200240, People's Republic of China
| | - Lin Xu
- Department of Gastroenterology, Shanghai Fifth People's Hospital, Fudan University, 128 Ruili Road, Shanghai 200240, People's Republic of China
| | - Nuo Cheng
- Department of Gastroenterology, Shanghai Fifth People's Hospital, Fudan University, 128 Ruili Road, Shanghai 200240, People's Republic of China
| | - Yan Chu
- Department of Gastroenterology, Shanghai Fifth People's Hospital, Fudan University, 128 Ruili Road, Shanghai 200240, People's Republic of China
| | - Hongwei Lyu
- Department of Gastroenterology, Shanghai Fifth People's Hospital, Fudan University, 128 Ruili Road, Shanghai 200240, People's Republic of China
| | - Fengyuan Chen
- Department of Gastroenterology, Shanghai Fifth People's Hospital, Fudan University, 128 Ruili Road, Shanghai 200240, People's Republic of China.
| |
Collapse
|
15
|
Abstract
Inflammatory cytokines are necessary for an acute response to injury and the progressive healing process. However, when this acute response does not resolve and becomes chronic, the same proteins that once promoted healing then contribute to chronic inflammatory pathologies, such as atherosclerosis. OPN (Osteopontin) is a secreted matricellular cytokine that signals through integrin and CD44 receptors, is highly upregulated in acute and chronic inflammatory settings, and has been implicated in physiological and pathophysiologic processes. Evidence from the literature suggests that OPN may fit within the Goldilocks paradigm with respect to cardiovascular disease, where acute increases are protective, attenuate vascular calcification, and promote postischemic neovascularization. In contrast, chronic increases in OPN are clinically associated with an increased risk for a major adverse cardiovascular event, and OPN expression is a strong predictor of cardiovascular disease independent of traditional risk factors. With the recent finding that humans express multiple OPN isoforms as the result of alternative splicing and that these isoforms have distinct biologic functions, future studies are required to determine what OPN isoform(s) are expressed in the setting of vascular disease and what role each of these isoforms plays in vascular disease progression. This review aims to discuss our current understanding of the role(s) of OPN in vascular disease pathologies using evidence from in vitro, animal, and clinical studies. Where possible, we discuss what is known about OPN isoform expression and our understanding of OPN isoform contributions to cardiovascular disease pathologies.
Collapse
Affiliation(s)
- Zoe Shin Yee Lok
- Department of Surgery, School of Clinical Sciences, Monash Health, Clayton, Australia (Z.S.Y.L.)
| | - Alicia N Lyle
- From the Division of Cardiology, Department of Medicine, Emory University School of Medicine, Atlanta, GA (A.N.L.)
| |
Collapse
|
16
|
Katholnig K, Schütz B, Fritsch SD, Schörghofer D, Linke M, Sukhbaatar N, Matschinger JM, Unterleuthner D, Hirtl M, Lang M, Herac M, Spittler A, Bergthaler A, Schabbauer G, Bergmann M, Dolznig H, Hengstschläger M, Magnuson MA, Mikula M, Weichhart T. Inactivation of mTORC2 in macrophages is a signature of colorectal cancer that promotes tumorigenesis. JCI Insight 2019; 4:124164. [PMID: 31619583 PMCID: PMC6824305 DOI: 10.1172/jci.insight.124164] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2018] [Accepted: 09/06/2019] [Indexed: 12/30/2022] Open
Abstract
The mechanistic target of rapamycin complex 2 (mTORC2) is a potentially novel and promising anticancer target due to its critical roles in proliferation, apoptosis, and metabolic reprogramming of cancer cells. However, the activity and function of mTORC2 in distinct cells within malignant tissue in vivo is insufficiently explored. Surprisingly, in primary human and mouse colorectal cancer (CRC) samples, mTORC2 signaling could not be detected in tumor cells. In contrast, only macrophages in tumor-adjacent areas showed mTORC2 activity, which was downregulated in stromal macrophages residing within human and mouse tumor tissues. Functionally, inhibition of mTORC2 by specific deletion of Rictor in macrophages stimulated tumorigenesis in a colitis-associated CRC mouse model. This phenotype was driven by a proinflammatory reprogramming of mTORC2-deficient macrophages that promoted colitis via the cytokine SPP1/osteopontin to stimulate tumor growth. In human CRC patients, high SPP1 levels and low mTORC2 activity in tumor-associated macrophages correlated with a worsened clinical prognosis. Treatment of mice with a second-generation mTOR inhibitor that inhibits mTORC2 and mTORC1 exacerbated experimental colorectal tumorigenesis in vivo. In conclusion, mTORC2 activity is confined to macrophages in CRC and limits tumorigenesis. These results suggest activation but not inhibition of mTORC2 as a therapeutic strategy for colitis-associated CRC.
Collapse
Affiliation(s)
- Karl Katholnig
- Center of Pathobiochemistry and Genetics, Institute of Medical Genetics
| | - Birgit Schütz
- Center of Pathobiochemistry and Genetics, Institute of Medical Genetics
| | | | - David Schörghofer
- Center of Pathobiochemistry and Genetics, Institute of Medical Genetics
| | - Monika Linke
- Center of Pathobiochemistry and Genetics, Institute of Medical Genetics
| | | | | | | | - Martin Hirtl
- Center of Pathobiochemistry and Genetics, Institute of Medical Genetics
| | - Michaela Lang
- Department of Internal Medicine III, Division of Gastroenterology and Hepatology
| | | | - Andreas Spittler
- Core Facility Flow Cytometry & Surgical Research Laboratories, Medical University of Vienna, Vienna, Austria
| | - Andreas Bergthaler
- CeMM Research Center for Molecular Medicine, Austrian Academy of Sciences, Vienna, Austria
| | - Gernot Schabbauer
- Institute for Physiology, Center for Physiology and Pharmacology, and
| | - Michael Bergmann
- Division of General Surgery, Department of Surgery, Medical University of Vienna, Vienna, Austria
| | - Helmut Dolznig
- Center of Pathobiochemistry and Genetics, Institute of Medical Genetics
| | | | - Mark A Magnuson
- Department of Molecular Physiology and Biophysics and Center for Stem Cell Biology, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - Mario Mikula
- Center of Pathobiochemistry and Genetics, Institute of Medical Genetics
| | - Thomas Weichhart
- Center of Pathobiochemistry and Genetics, Institute of Medical Genetics
| |
Collapse
|
17
|
Nazmi A, Hoek KL, Greer MJ, Piazuelo MB, Minato N, Olivares-Villagómez D. Innate CD8αα+ cells promote ILC1-like intraepithelial lymphocyte homeostasis and intestinal inflammation. PLoS One 2019; 14:e0215883. [PMID: 31291255 PMCID: PMC6619599 DOI: 10.1371/journal.pone.0215883] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2019] [Accepted: 06/27/2019] [Indexed: 12/22/2022] Open
Abstract
Innate CD8αα+ cells, also referred to as iCD8α cells, are TCR-negative intraepithelial lymphocytes (IEL) possessing cytokine and chemokine profiles and functions related to innate immune cells. iCD8α cells constitute an important source of osteopontin in the intestinal epithelium. Osteopontin is a pleiotropic cytokine with diverse roles in bone and tissue remodeling, but also has relevant functions in the homeostasis of immune cells. In this report, we present evidence for the role of iCD8α cells in the homeostasis of TCR-negative NKp46+NK1.1+ IEL (ILC1-like). We also show that the effect of iCD8α cells on ILC1-like IEL is enhanced in vitro by osteopontin. We show that in the absence of iCD8α cells, the number of NKp46+NK1.1+ IEL is significantly reduced. These ILC1-like cells are involved in intestinal pathogenesis in the anti-CD40 mouse model of intestinal inflammation. Reduced iCD8α cell numbers results in a milder form of intestinal inflammation in this disease model, whereas treatment with osteopontin increases disease severity. Collectively, our results suggest that iCD8α cells promote survival of NKp46+NK1.1+ IEL, which significantly impacts the development of intestinal inflammation.
Collapse
Affiliation(s)
- Ali Nazmi
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee, United States of America
| | - Kristen L. Hoek
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee, United States of America
| | - Michael J. Greer
- Department of Biomedical Informatics, Vanderbilt University, Nashville, Tennessee, United States of America
| | - Maria B. Piazuelo
- Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, United States of America
| | - Nagahiro Minato
- Medical Innovation Center, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Danyvid Olivares-Villagómez
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee, United States of America
- Vanderbilt Institute for Infection, Immunology and Inflammation, Vanderbilt University Medical Center, Nashville, Tennessee, United States of America
| |
Collapse
|
18
|
Rowley AT, Nagalla RR, Wang S, Liu WF. Extracellular Matrix-Based Strategies for Immunomodulatory Biomaterials Engineering. Adv Healthc Mater 2019; 8:e1801578. [PMID: 30714328 DOI: 10.1002/adhm.201801578] [Citation(s) in RCA: 109] [Impact Index Per Article: 21.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2018] [Revised: 01/08/2019] [Indexed: 12/14/2022]
Abstract
The extracellular matrix (ECM) is a complex and dynamic structural scaffold for cells within tissues and plays an important role in regulating cell function. Recently it has become appreciated that the ECM contains bioactive motifs that can directly modulate immune responses. This review describes strategies for engineering immunomodulatory biomaterials that utilize natural ECM-derived molecules and have the potential to harness the immune system for applications ranging from tissue regeneration to drug delivery. A top-down approach utilizes full-length ECM proteins, including collagen, fibrin, or hyaluronic acid-based materials, as well as matrices derived from decellularized tissue. These materials have the benefit of maintaining natural conformation and structure but are often heterogeneous and encumber precise control. By contrast, a bottom-up approach leverages immunomodulatory domains, such as Arg-Gly-Asp (RGD), matrix metalloproteinase (MMP)-sensitive peptides, or leukocyte-associated immunoglobulin-like receptor-1(LAIR-1) ligands, by incorporating them into synthetic materials. These materials have tunable control over immune cell functions and allow for combinatorial approaches. However, the synthetic approach lacks the full natural context of the original ECM protein. These two approaches provide a broad range of engineering techniques for immunomodulation through material interactions and hold the potential for the development of future therapeutic applications.
Collapse
Affiliation(s)
- Andrew T. Rowley
- Department of Chemical and Biomolecular EngineeringUniversity of California Irvine CA 92697 USA
| | - Raji R. Nagalla
- Department of Biomedical EngineeringUniversity of California Irvine CA 92697 USA
| | - Szu‐Wen Wang
- Department of Chemical and Biomolecular EngineeringUniversity of California Irvine CA 92697 USA
- Department of Biomedical EngineeringUniversity of California Irvine CA 92697 USA
- Department of Materials Science and EngineeringUniversity of California Irvine CA 92697 USA
| | - Wendy F. Liu
- Department of Chemical and Biomolecular EngineeringUniversity of California Irvine CA 92697 USA
- Department of Biomedical EngineeringUniversity of California Irvine CA 92697 USA
- The Edwards Lifesciences Center for Advanced Cardiovascular TechnologyUniversity of California Irvine CA 92697 USA
| |
Collapse
|
19
|
OPN gene locus is associated with the risk of knee osteoarthritis: a case-control study. Biosci Rep 2019; 39:BSR20182023. [PMID: 30777930 PMCID: PMC6393225 DOI: 10.1042/bsr20182023] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2018] [Revised: 01/29/2019] [Accepted: 02/12/2019] [Indexed: 12/13/2022] Open
Abstract
Background/aims: Studies have demonstrated that osteopontin (OPN) was associated with the severity and development of knee osteoarthritis (OA). Methods: The purpose of this case-control study was to investigate the association between OPN gene rs11730582 polymorphism and knee OA risk in a Chinese population. Genotyping was analyzed using standard PCR and restriction fragment length polymorphism (PCR-RFLP). Results: The present study found that C allele or CC genotype of OPN gene rs11730582 polymorphism was related to decreased risk for knee OA. Furthermore, positive associations were obtained amongst the females, and body mass index (BMI) < 25 kg/m2 groups. Conclusions: To sum up, the present study reveals that OPN gene rs11730582 polymorphism decreases the risk of knee OA in Chinese Han population.
Collapse
|
20
|
Woo SH, Lee SH, Park JW, Go DM, Kim DY. Osteopontin Protects Colonic Mucosa from Dextran Sodium Sulfate-Induced Acute Colitis in Mice by Regulating Junctional Distribution of Occludin. Dig Dis Sci 2019; 64:421-431. [PMID: 30146676 DOI: 10.1007/s10620-018-5246-6] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/23/2018] [Accepted: 08/09/2018] [Indexed: 02/08/2023]
Abstract
BACKGROUND Osteopontin (OPN) has been reported to play an important role in intestinal mucosal protection. Although OPN may have positive effects on tight junctions, the exact relationship between OPN and tight junctions has yet to be elucidated. AIMS To investigate the role of OPN on tight junctions. METHODS We evaluated clinical signs and histopathology of acute colitis induced by dextran sodium sulfate (DSS) in OPN knockout and wild-type (WT) mice in vivo. Expression levels of occludin and zonula occludens-1 were examined using immunofluorescence. For in vitro analysis, an siRNA-mediated OPN-suppressed Caco-2 monolayer was used. Expression levels and patterns of occludin were analyzed by immunofluorescence, and transepithelial electrical resistance (TER) was measured to evaluate barrier function. Triton X-100 fractionation was used to analyze phosphorylated occludin associated with tight junctional localization. RESULTS OPN deficiency resulted in an elevated disease activity index, shortened colon length, and aggravated histological signs in mice with DSS-induced acute colitis compared to WT mice. OPN deficiency decreased occludin expression in the colonic mucosa. In Caco-2 monolayers, OPN suppression reduced junctional occludin and redistributed it into the intracellular compartment with decreased TER. Furthermore, western blot for occludin from Triton X-100 insoluble fraction revealed that OPN suppression reduced the phosphorylated form of occludin, which is actually distributed in the tight junction. CONCLUSIONS Our study showed that OPN is essential for maintaining the tight junction complex by allowing occludin to localize at tight junctions. This could constitute additional evidence that OPN plays a crucial role in intestinal mucosal protection.
Collapse
Affiliation(s)
- Sang-Ho Woo
- Department of Veterinary Pathology, College of Veterinary Medicine, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul, 08826, South Korea
| | - Su-Hyung Lee
- Branch of Carcinogenesis and Metastasis, Research Institute of National Cancer Center, Goyang, Gyeonggi, 10408, South Korea
| | - Jun-Won Park
- Division of Genetics, Cancer Research Institute, Kanazawa University, Kanazawa, 920-1192, Japan
| | - Du-Min Go
- Department of Veterinary Pathology, College of Veterinary Medicine, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul, 08826, South Korea
| | - Dae-Yong Kim
- Department of Veterinary Pathology, College of Veterinary Medicine, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul, 08826, South Korea.
| |
Collapse
|
21
|
Is Osteopontin a Friend or Foe of Cell Apoptosis in Inflammatory Gastrointestinal and Liver Diseases? Int J Mol Sci 2017; 19:ijms19010007. [PMID: 29267211 PMCID: PMC5795959 DOI: 10.3390/ijms19010007] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2017] [Revised: 12/11/2017] [Accepted: 12/19/2017] [Indexed: 12/15/2022] Open
Abstract
Osteopontin (OPN) is involved in a variety of biological processes, including bone remodeling, innate immunity, acute and chronic inflammation, and cancer. The expression of OPN occurs in various tissues and cells, including intestinal epithelial cells and immune cells such as macrophages, dendritic cells, and T lymphocytes. OPN plays an important role in the efficient development of T helper 1 immune responses and cell survival by inhibiting apoptosis. The association of OPN with apoptosis has been investigated. In this review, we described the role of OPN in inflammatory gastrointestinal and liver diseases, focusing on the association of OPN with apoptosis. OPN changes its association with apoptosis depending on the type of disease and the phase of disease activity, acting as a promoter or a suppressor of inflammation and inflammatory carcinogenesis. It is essential that the roles of OPN in those diseases are elucidated, and treatments based on its mechanism are developed.
Collapse
|
22
|
Kim ID, Lee H, Jin YC, Lee JK. Osteopontin Peptide Icosamer Containing RGD and SLAYGLR Motifs Enhances the Motility and Phagocytic Activity of Microglia. Exp Neurobiol 2017; 26:339-349. [PMID: 29302201 PMCID: PMC5746499 DOI: 10.5607/en.2017.26.6.339] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2017] [Revised: 11/16/2017] [Accepted: 12/01/2017] [Indexed: 11/19/2022] Open
Abstract
Osteopontin (OPN) is a secreted glycoprotein that is expressed in various tissues, including brain, and mediates a wide range of cellular activities. In a previous study, the authors observed the robust neuroprotective effects of recombinant OPN and of RGD and SLAYGLR-containing OPN-peptide icosamer (OPNpt20) in an animal model of transient focal ischemia, and demonstrated anti-inflammatory and pro-angiogenic effects of OPNpt20 in the postischemic brain. In the present study, we investigated the effects of OPNpt20 on the motility and phagocytic activity of BV2 cells (a microglia cell line). F-actin polymerization and cell motility were significantly enhanced in OPNpt20-treated BV2 cells, and numbers of filopodia-like processes increased and lamellipodia-like structures enlarged and thickened. In addition, treatment of cells with either of three mutant OPN icosamers containing mutation within RGD, SLAY, or RGDSLAY showed that the RGD and SLAY motifs of OPNpt20 play critical roles in the enhancement of cell motility, and the interaction between exogenous OPNpt20 and endogenous αv and α4 integrin and the activations of FAK, Erk, and Akt signaling pathways were found to be involved in the OPNpt20-mediated induction of cell motility. Furthermore, phagocytic activity of microglia was also significantly enhanced by OPNpt20 in a RGD and SLAY dependent manner. These results indicate OPNpt20 containing RGD and SLAY motifs triggers microglial motility and phagocytic activity and OPNpt20-integrin mediated signaling plays a critical role in these activities.
Collapse
Affiliation(s)
- Il-Doo Kim
- Department of Anatomy, Inha University School of Medicine, Incheon, Korea.,Medical Research Center, Inha University School of Medicine, Incheon, Korea
| | - Hahnbie Lee
- Department of Anatomy, Inha University School of Medicine, Incheon, Korea.,Medical Research Center, Inha University School of Medicine, Incheon, Korea
| | - Yin-Chuan Jin
- Department of Histology and Embryology, Binzhou Medical University, Yantai 264000, China
| | - Ja-Kyeong Lee
- Department of Anatomy, Inha University School of Medicine, Incheon, Korea.,Medical Research Center, Inha University School of Medicine, Incheon, Korea
| |
Collapse
|
23
|
Osteopontin and the immune system: another brick in the wall. Cell Mol Immunol 2017; 15:405-407. [PMID: 28967877 DOI: 10.1038/cmi.2017.94] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2017] [Accepted: 08/06/2017] [Indexed: 12/21/2022] Open
|
24
|
Skewing of the population balance of lymphoid and myeloid cells by secreted and intracellular osteopontin. Nat Immunol 2017; 18:973-984. [PMID: 28671690 PMCID: PMC5568448 DOI: 10.1038/ni.3791] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2017] [Accepted: 06/13/2017] [Indexed: 12/12/2022]
Abstract
The balance of myeloid populations and lymphoid populations must be well controlled. Here we found that osteopontin (OPN) skewed this balance during pathogenic conditions such as infection and autoimmunity. Notably, two isoforms of OPN exerted distinct effects in shifting this balance through cell-type-specific regulation of apoptosis. Intracellular OPN (iOPN) diminished the population size of myeloid progenitor cells and myeloid cells, and secreted OPN (sOPN) increase the population size of lymphoid cells. The total effect of OPN on skewing the leukocyte population balance was observed as host sensitivity to early systemic infection with Candida albicans and T cell-mediated colitis. Our study suggests previously unknown detrimental roles for two OPN isoforms in causing the imbalance of leukocyte populations.
Collapse
|
25
|
Osteopontin Deficiency Suppresses Intestinal Tumor Development in Apc-Deficient Min Mice. Int J Mol Sci 2017; 18:ijms18051058. [PMID: 28505114 PMCID: PMC5454970 DOI: 10.3390/ijms18051058] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2017] [Revised: 05/05/2017] [Accepted: 05/09/2017] [Indexed: 12/18/2022] Open
Abstract
Osteopontin (OPN) is a secreted phosphoglycoprotein, and is a transcriptional target of aberrant Wnt signaling. OPN is upregulated in human colon cancers, and is suggested to enhance cancer progression. In this study, the effect of deficiency of OPN on intestinal tumor development in Apc-deficient Min mice was investigated. At 16 weeks of age, the number of small intestinal polyps in Min/OPN(+/−) and Min/OPN(−/−) mice was lower than that of Min/OPN(+/+) mice. Colorectal tumor incidences and multiplicities in Min/OPN(+/−) and Min/OPN(−/−) mice were significantly lower than those in Min/OPN(+/+) mice, being 48% and 0.6 ± 0.8, 50% and 0.8 ± 0.9 vs. 80% and 1.6 ± 1.7, respectively. OPN expression in colorectal tumors was strongly upregulated in Min/OPN(+/+) compared to adjacent non-tumor parts, but was decreased in Min/OPN(+/−) and not detected in Min/OPN(−/−). Targets of OPN, matrix metalloproteinases (MMPs)-3, -9, and -13 were lowered by OPN deficiency. Macrophage marker F4/80 in colorectal tumors was also lowered by OPN deficiency. MMP-9 expression was observed in tumor cells and tumor-infiltrating neutrophils. These results indicate that induction of OPN by aberrant Wnt signaling could enhance colorectal tumor development in part by upregulation of MMP-3, -9, and -13 and infiltration of macrophage and neutrophils. Suppression of OPN expression could contribute to tumor prevention, but complete deficiency of OPN may cause some adverse effects.
Collapse
|
26
|
Ito K, Nakajima A, Fukushima Y, Suzuki K, Sakamoto K, Hamazaki Y, Ogasawara K, Minato N, Hattori M. The potential role of Osteopontin in the maintenance of commensal bacteria homeostasis in the intestine. PLoS One 2017; 12:e0173629. [PMID: 28296922 PMCID: PMC5351998 DOI: 10.1371/journal.pone.0173629] [Citation(s) in RCA: 12] [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: 09/01/2016] [Accepted: 02/22/2017] [Indexed: 01/28/2023] Open
Abstract
Osteopontin (Opn), a multifunctional extracellular matrix protein, is implicated in the pathogenesis of various inflammatory disorders. Under physiologic conditions, its expression is restricted to certain tissues including bone and kidney tubule. However, cellular activation during disease development induces Opn expression in various immune cells. In this study, using Opn-EGFP knock-in (KI) mice we found that CD8α+ T cells in the intestinal tissues, including Peyer’s patch, lamina propria and epithelium, express Opn under steady state conditions. Therefore, we examined the role of Opn-expressing CD8α+ T cells in intestinal homeostasis. Interestingly, Opn knockout (KO) mice had altered fecal microflora concordant with a reduction of TCRγδ+ intraepithelial lymphocytes (IELs). Consistent with this result, both treatment with anti-Opn blocking antibody and deficiency of Opn resulted in decreased survival of TCRγδ+ and TCRαβ+ IELs. This data suggests that a possibility that Opn may function as a survival factor for IELs in the intestinal tissue. Collectively, these data suggest the possibility that Opn might regulate the homeostasis of intestinal microflora through maintenance of TCRγδ+ IELs, possibly by support of IEL survival.
Collapse
Affiliation(s)
- Koyu Ito
- Center for Innovation in Immunoregulative Technology and Therapeutics, Graduate School of Medicine, Kyoto University, Yoshida Konoe-cho, Sakyo-ku, Kyoto, Japan
- Department of Immunobiology, Institute of Development, Ageing, and Cancer, Tohoku University, Aoba-ku, Sendai, Miyagi, Japan
- * E-mail: (KI); (MH)
| | - Akira Nakajima
- Center for Innovation in Immunoregulative Technology and Therapeutics, Graduate School of Medicine, Kyoto University, Yoshida Konoe-cho, Sakyo-ku, Kyoto, Japan
| | - Yuji Fukushima
- Center for Innovation in Immunoregulative Technology and Therapeutics, Graduate School of Medicine, Kyoto University, Yoshida Konoe-cho, Sakyo-ku, Kyoto, Japan
| | - Keiichiro Suzuki
- Center for Innovation in Immunoregulative Technology and Therapeutics, Graduate School of Medicine, Kyoto University, Yoshida Konoe-cho, Sakyo-ku, Kyoto, Japan
| | - Keiko Sakamoto
- Department of Immunology and Cell Biology, Graduate School of Medicine, Kyoto University, Yoshida Konoe-cho, Sakyo-ku, Kyoto, Japan
| | - Yoko Hamazaki
- Department of Immunology and Cell Biology, Graduate School of Medicine, Kyoto University, Yoshida Konoe-cho, Sakyo-ku, Kyoto, Japan
| | - Kouetsu Ogasawara
- Department of Immunobiology, Institute of Development, Ageing, and Cancer, Tohoku University, Aoba-ku, Sendai, Miyagi, Japan
| | - Nagahiro Minato
- Department of Immunology and Cell Biology, Graduate School of Medicine, Kyoto University, Yoshida Konoe-cho, Sakyo-ku, Kyoto, Japan
| | - Masakazu Hattori
- Center for Innovation in Immunoregulative Technology and Therapeutics, Graduate School of Medicine, Kyoto University, Yoshida Konoe-cho, Sakyo-ku, Kyoto, Japan
- * E-mail: (KI); (MH)
| |
Collapse
|
27
|
Kawakami K, Minami N, Matsuura M, Iida T, Toyonaga T, Nagaishi K, Arimura Y, Fujimiya M, Uede T, Nakase H. Osteopontin attenuates acute gastrointestinal graft-versus-host disease by preventing apoptosis of intestinal epithelial cells. Biochem Biophys Res Commun 2017; 485:468-475. [PMID: 28192120 DOI: 10.1016/j.bbrc.2017.02.047] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2017] [Accepted: 02/08/2017] [Indexed: 11/16/2022]
Abstract
BACKGROUND AND AIMS Acute graft-versus-host disease (GVHD) is a major complication after allogeneic hematopoietic stem cell transplantation, which often targets gastrointestinal (GI) tract. Osteopontin (OPN) plays an important physiological role in the efficient development of Th1 immune responses and cell survival by inhibiting apoptosis. The role of OPN in acute GI-GVHD is poorly understood. In the present study, we investigated the role of OPN in donor T cells in the pathogenicity of acute GI-GVHD. METHODS OPN knockout (KO) mice and C57BL/6 (B6) mice were used as donors, and (C57BL/6 × DBA/2) F1 (BDF1) mice were used as allograft recipients. Mice with acute GI-GVHD were divided into three groups: the control group (BDF1→BDF1), B6 group (B6→BDF1), and OPN-KO group (OPN-KO→BDF1). Bone marrow cells and spleen cells from donors were transplanted to lethally irradiated recipients. Clinical GVHD scores were assessed daily. Recipients were euthanized on day 7 after transplantation, and colons and small intestines were collected for various analyses. RESULTS The clinical GVHD score in the OPN-KO group was significantly increased compared with the B6 and control groups. We observed a difference in the severity of colonic GVHD between the OPN-KO group and B6 group, but not small intestinal-GVHD between these groups. Interferon-γ, Tumor necrosis factor-α, Interleukin-17A, and Interleukin-18 gene expression in the OPN-KO group was differed between the colon and small intestine. Flow cytometric analysis revealed that the fluorescence intensity of splenic and colonic CD8 T cells expressing Fas Ligand was increased in the OPN-KO group compared with the B6 group. CONCLUSION We demonstrated that the importance of OPN in T cells in the onset of acute GI-GVHD involves regulating apoptosis of the intestinal cell via the Fas-Fas Ligand pathway.
Collapse
Affiliation(s)
- Kentaro Kawakami
- Department of Gastroenterology and Hepatology, Sapporo Medical University, School of Medicine, Minami 1-jo Nishi 17-chome, Chuo-ku, Sapporo, Hokkaido 060-8556, Japan
| | - Naoki Minami
- Department of Gastroenterology and Hepatology, Kyoto University, Graduate School of Medicine, 54 Shogoin, Kawahara-Cho, Sakyo-ku, Kyoto 606-8507, Japan
| | - Minoru Matsuura
- Department of Gastroenterology and Hepatology, Kyoto University, Graduate School of Medicine, 54 Shogoin, Kawahara-Cho, Sakyo-ku, Kyoto 606-8507, Japan
| | - Tomoya Iida
- Department of Gastroenterology and Hepatology, Sapporo Medical University, School of Medicine, Minami 1-jo Nishi 17-chome, Chuo-ku, Sapporo, Hokkaido 060-8556, Japan
| | - Takahiko Toyonaga
- Center for Advanced IBD Research and Treatment, Kitasato University Kitasato Institute Hospital, 5-9-1, Shirokane, Minato-ku, Tokyo 108-8642, Japan
| | - Kanna Nagaishi
- Department of Anatomy, Sapporo Medical University, School of Medicine, Minami 1-jo Nishi 17-chome, Chuo-ku, Sapporo, Hokkaido 060-8556, Japan
| | - Yoshiaki Arimura
- Department of Gastroenterology and Hepatology, Sapporo Medical University, School of Medicine, Minami 1-jo Nishi 17-chome, Chuo-ku, Sapporo, Hokkaido 060-8556, Japan
| | - Mineko Fujimiya
- Department of Anatomy, Sapporo Medical University, School of Medicine, Minami 1-jo Nishi 17-chome, Chuo-ku, Sapporo, Hokkaido 060-8556, Japan
| | - Toshimitsu Uede
- Department of Matrix Medicine, Institute for Genetic Medicine, Hokkaido University, Sapporo Kita 15-jo Nishi 7-chome, Kita-ku, Sapporo, Hokkaido 060-0815, Japan
| | - Hiroshi Nakase
- Department of Gastroenterology and Hepatology, Sapporo Medical University, School of Medicine, Minami 1-jo Nishi 17-chome, Chuo-ku, Sapporo, Hokkaido 060-8556, Japan.
| |
Collapse
|
28
|
Clemente N, Raineri D, Cappellano G, Boggio E, Favero F, Soluri MF, Dianzani C, Comi C, Dianzani U, Chiocchetti A. Osteopontin Bridging Innate and Adaptive Immunity in Autoimmune Diseases. J Immunol Res 2016; 2016:7675437. [PMID: 28097158 PMCID: PMC5206443 DOI: 10.1155/2016/7675437] [Citation(s) in RCA: 103] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2016] [Revised: 10/02/2016] [Accepted: 10/19/2016] [Indexed: 12/21/2022] Open
Abstract
Osteopontin (OPN) regulates the immune response at multiple levels. Physiologically, it regulates the host response to infections by driving T helper (Th) polarization and acting on both innate and adaptive immunity; pathologically, it contributes to the development of immune-mediated and inflammatory diseases. In some cases, the mechanisms of these effects have been described, but many aspects of the OPN function remain elusive. This is in part ascribable to the fact that OPN is a complex molecule with several posttranslational modifications and it may act as either an immobilized protein of the extracellular matrix or a soluble cytokine or an intracytoplasmic molecule by binding to a wide variety of molecules including crystals of calcium phosphate, several cell surface receptors, and intracytoplasmic molecules. This review describes the OPN structure, isoforms, and functions and its role in regulating the crosstalk between innate and adaptive immunity in autoimmune diseases.
Collapse
Affiliation(s)
- Nausicaa Clemente
- Department of Health Sciences and Interdisciplinary Research Center of Autoimmune Diseases (IRCAD), “A. Avogadro” University of Piemonte Orientale (UPO), Novara, Italy
| | - Davide Raineri
- Department of Health Sciences and Interdisciplinary Research Center of Autoimmune Diseases (IRCAD), “A. Avogadro” University of Piemonte Orientale (UPO), Novara, Italy
| | - Giuseppe Cappellano
- Biocenter, Division for Experimental Pathophysiology and Immunology, Laboratory of Autoimmunity, Medical University of Innsbruck, Innsbruck, Austria
| | - Elena Boggio
- Department of Health Sciences and Interdisciplinary Research Center of Autoimmune Diseases (IRCAD), “A. Avogadro” University of Piemonte Orientale (UPO), Novara, Italy
| | - Francesco Favero
- Department of Health Sciences and Interdisciplinary Research Center of Autoimmune Diseases (IRCAD), “A. Avogadro” University of Piemonte Orientale (UPO), Novara, Italy
| | - Maria Felicia Soluri
- Department of Health Sciences and Interdisciplinary Research Center of Autoimmune Diseases (IRCAD), “A. Avogadro” University of Piemonte Orientale (UPO), Novara, Italy
| | - Chiara Dianzani
- Department of Drug Science and Technology, University of Torino, Torino, Italy
| | - Cristoforo Comi
- Department of Translational Medicine, Neurology Unit, “A. Avogadro” UPO, Novara, Italy
| | - Umberto Dianzani
- Department of Health Sciences and Interdisciplinary Research Center of Autoimmune Diseases (IRCAD), “A. Avogadro” University of Piemonte Orientale (UPO), Novara, Italy
| | - Annalisa Chiocchetti
- Department of Health Sciences and Interdisciplinary Research Center of Autoimmune Diseases (IRCAD), “A. Avogadro” University of Piemonte Orientale (UPO), Novara, Italy
| |
Collapse
|
29
|
Melve GK, Ersvaer E, Akkök ÇA, Ahmed AB, Kristoffersen EK, Hervig T, Bruserud Ø. Immunomodulation Induced by Stem Cell Mobilization and Harvesting in Healthy Donors: Increased Systemic Osteopontin Levels after Treatment with Granulocyte Colony-Stimulating Factor. Int J Mol Sci 2016; 17:ijms17071158. [PMID: 27447610 PMCID: PMC4964530 DOI: 10.3390/ijms17071158] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2016] [Revised: 06/25/2016] [Accepted: 07/11/2016] [Indexed: 12/13/2022] Open
Abstract
Peripheral blood stem cells from healthy donors mobilized by granulocyte colony-stimulating factor (G-CSF) and harvested by leukapheresis are commonly used for allogeneic stem cell transplantation. The frequency of severe graft versus host disease is similar for patients receiving peripheral blood and bone marrow allografts, even though the blood grafts contain more T cells, indicating mobilization-related immunoregulatory effects. The regulatory phosphoprotein osteopontin was quantified in plasma samples from healthy donors before G-CSF treatment, after four days of treatment immediately before and after leukapheresis, and 18–24 h after apheresis. Myeloma patients received chemotherapy, combined with G-CSF, for stem cell mobilization and plasma samples were prepared immediately before, immediately after, and 18–24 h after leukapheresis. G-CSF treatment of healthy stem cell donors increased plasma osteopontin levels, and a further increase was seen immediately after leukapheresis. The pre-apheresis levels were also increased in myeloma patients compared to healthy individuals. Finally, in vivo G-CSF exposure did not alter T cell expression of osteopontin ligand CD44, and in vitro osteopontin exposure induced only small increases in anti-CD3- and anti-CD28-stimulated T cell proliferation. G-CSF treatment, followed by leukapheresis, can increase systemic osteopontin levels, and this effect may contribute to the immunomodulatory effects of G-CSF treatment.
Collapse
Affiliation(s)
- Guro Kristin Melve
- Department of Immunology and Transfusion Medicine, Haukeland University Hospital, N-5021 Bergen, Norway.
- Department of Clinical Science, University of Bergen, N-5020 Bergen, Norway.
| | - Elisabeth Ersvaer
- Department of Biomedical Laboratory Sciences and Chemical Engineering, Faculty of Engineering and Business Administration, Bergen University College, N-5020 Bergen, Norway.
| | - Çiğdem Akalın Akkök
- Department of Immunology and Transfusion Medicine, Oslo University Hospital, Ullevål, N-0424 Oslo, Norway.
| | - Aymen Bushra Ahmed
- Division for Hematology, Department of Medicine, Haukeland University Hospital, N-5021 Bergen, Norway.
| | - Einar K Kristoffersen
- Department of Immunology and Transfusion Medicine, Haukeland University Hospital, N-5021 Bergen, Norway.
- Department of Clinical Science, University of Bergen, N-5020 Bergen, Norway.
| | - Tor Hervig
- Department of Immunology and Transfusion Medicine, Haukeland University Hospital, N-5021 Bergen, Norway.
- Department of Clinical Science, University of Bergen, N-5020 Bergen, Norway.
| | - Øystein Bruserud
- Department of Clinical Science, University of Bergen, N-5020 Bergen, Norway.
- Division for Hematology, Department of Medicine, Haukeland University Hospital, N-5021 Bergen, Norway.
| |
Collapse
|
30
|
Nagy Z, Acs B, Butz H, Feldman K, Marta A, Szabo PM, Baghy K, Pazmany T, Racz K, Liko I, Patocs A. Overexpression of GRß in colonic mucosal cell line partly reflects altered gene expression in colonic mucosa of patients with inflammatory bowel disease. J Steroid Biochem Mol Biol 2016; 155:76-84. [PMID: 26480216 DOI: 10.1016/j.jsbmb.2015.10.006] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/04/2015] [Revised: 09/11/2015] [Accepted: 10/07/2015] [Indexed: 10/22/2022]
Abstract
The glucocorticoid receptor (GR) plays a crucial role in inflammatory responses. GR has several isoforms, of which the most deeply studied are the GRα and GRß. Recently it has been suggested that in addition to its negative dominant effect on GRα, the GRß may have a GRα-independent transcriptional activity. The GRß isoform was found to be frequently overexpressed in various autoimmune diseases, including inflammatory bowel disease (IBD). In this study, we wished to test whether the gene expression profile found in a GRß overexpressing intestinal cell line (Caco-2GRß) might mimic the gene expression alterations found in patients with IBD. Whole genome microarray analysis was performed in both normal and GRß overexpressing Caco-2 cell lines with and without dexamethasone treatment. IBD-related genes were identified from a meta-analysis of 245 microarrays available in online microarray deposits performed on intestinal mucosa samples from patients with IBD and healthy individuals. The differentially expressed genes were further studied using in silico pathway analysis. Overexpression of GRß altered a large proportion of genes that were not regulated by dexamethasone suggesting that GRß may have a GRα-independent role in the regulation of gene expression. About 10% of genes differentially expressed in colonic mucosa samples from IBD patients compared to normal subjects were also detected in Caco-2 GRß intestinal cell line. Common genes are involved in cell adhesion and cell proliferation. Overexpression of GRß in intestinal cells may affect appropriate mucosal repair and intact barrier function. The proposed novel role of GRß in intestinal epithelium warrants further studies.
Collapse
Affiliation(s)
- Zsolt Nagy
- 2nd Department of Medicine, Faculty of Medicine, Semmelweis University, Budapest, Hungary; Hungarian Academy of Sciences-Semmelweis University "Lendulet" Hereditary Endocrine Tumors Research Group, Budapest, Hungary
| | - Bence Acs
- 2nd Department of Medicine, Faculty of Medicine, Semmelweis University, Budapest, Hungary
| | - Henriett Butz
- Hungarian Academy of Sciences-Semmelweis University "Lendulet" Hereditary Endocrine Tumors Research Group, Budapest, Hungary; Hungarian Academy of Sciences-Semmelweis University Molecular Medicine Research Group, Budapest, Hungary
| | - Karolina Feldman
- 2nd Department of Medicine, Faculty of Medicine, Semmelweis University, Budapest, Hungary
| | - Alexa Marta
- 2nd Department of Medicine, Faculty of Medicine, Semmelweis University, Budapest, Hungary
| | - Peter M Szabo
- Hungarian Academy of Sciences-Semmelweis University Molecular Medicine Research Group, Budapest, Hungary
| | - Kornelia Baghy
- 1st Department of Pathology and Experimental Cancer Research, Semmelweis University, Budapest, Hungary
| | | | - Karoly Racz
- 2nd Department of Medicine, Faculty of Medicine, Semmelweis University, Budapest, Hungary; Hungarian Academy of Sciences-Semmelweis University Molecular Medicine Research Group, Budapest, Hungary
| | - Istvan Liko
- Hungarian Academy of Sciences-Semmelweis University "Lendulet" Hereditary Endocrine Tumors Research Group, Budapest, Hungary; Gedeon Richter PLC, Budapest, Hungary
| | - Attila Patocs
- Hungarian Academy of Sciences-Semmelweis University "Lendulet" Hereditary Endocrine Tumors Research Group, Budapest, Hungary; Department of Laboratory Medicine, Semmelweis University, Budapest, Hungary.
| |
Collapse
|
31
|
Abstract
Since its initial identification as one of the genes most highly upregulated upon T-cell activation, osteopontin (or Eta-1, as it was designated then) has been demonstrated to have many roles in the regulation of the immune response on multiple levels. It contributes to the development of immune-mediated and inflammatory diseases, and it regulates the host response to infection. In some cases, the mechanisms of these effects have been elucidated, while other mechanistic functions of the protein remain obscure. The protein itself makes these analyses complex, since it binds to a series of different integrins, and in addition to its classically secreted form, an intracellular form of osteopontin has been identified, which participates in several aspects of immune regulation. In this review, we focus on the role of osteopontin in a series of immune-related diseases, particularly those where significant advances have been made in recent years: multiple sclerosis, rheumatoid arthritis, lupus and related diseases, Sjögren's disease, colitis, and 1 area of inflammatory pathology, alcoholic and nonalcoholic liver diseases. A recurring theme in these diseases is a link between osteopontin and pathogenic T cells, particularly T helper 17 cells, where osteopontin produced by dendritic cells supports IL-17 expression, contributing to pathology. In addition, a role for osteopontin in B-cell differentiation is becoming clear. In general, osteopontin contributes to pathology in these diseases, but there are examples where it has a protective role; deciphering the mechanisms underlying these differences and the specific receptors for osteopontin will be a research challenge for the future. Aside from its newly discovered role in the development of Sjögren's disease, the role of osteopontin in inflammatory conditions in the oral cavity is still poorly understood. Elucidation of this role will be of interest.
Collapse
Affiliation(s)
- S R Rittling
- The Forsyth Institute, Cambridge, MA, USA Harvard School of Dental Medicine, Boston, MA, USA
| | - R Singh
- The Forsyth Institute, Cambridge, MA, USA
| |
Collapse
|
32
|
Toyonaga T, Nakase H, Ueno S, Matsuura M, Yoshino T, Honzawa Y, Itou A, Namba K, Minami N, Yamada S, Koshikawa Y, Uede T, Chiba T, Okazaki K. Osteopontin Deficiency Accelerates Spontaneous Colitis in Mice with Disrupted Gut Microbiota and Macrophage Phagocytic Activity. PLoS One 2015; 10:e0135552. [PMID: 26274807 PMCID: PMC4537118 DOI: 10.1371/journal.pone.0135552] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2014] [Accepted: 07/24/2015] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND Osteopontin (OPN) is a multifunctional protein expressed in a variety of tissues and cells. Recent studies revealed increased OPN expression in the inflamed intestinal tissues of patients with inflammatory bowel disease (IBD). The role of OPN in the pathophysiology of IBD, however, remains unclear. AIMS To investigate the role of OPN in the development of intestinal inflammation using a murine model of IBD, interleukin-10 knock out (IL-10 KO) mice. METHODS We compared the development of colitis between IL-10 KO and OPN/IL-10 double KO (DKO) mice. OPN expression in the colonic tissues of IL-10 KO mice was examined by fluorescence in situ hybridization (FISH) analysis. Enteric microbiota were compared between IL-10 KO and OPN/IL-10 DKO mice by terminal restriction fragment length polymorphism analysis. The effect of OPN on macrophage phagocytic function was evaluated by phagocytosis assay. RESULTS OPN/IL-10 DKO mice had an accelerated onset of colitis compared to IL-10 KO mice. FISH analysis revealed enhanced OPN synthesis in the colonic epithelial cells of IL-10 KO mice. OPN/IL-10 DKO mice had a distinctly different enteric bacterial profile with a significantly lower abundance of Clostridium subcluster XIVa and a greater abundance of Clostridium cluster XVIII compared to IL-10 KO mice. Intracellular OPN deletion in macrophages impaired phagocytosis of fluorescence particle-conjugated Escherichia coli in vitro. Exogenous OPN enhanced phagocytosis by OPN-deleted macrophages when administered at doses of 1 to 100 ng/ml, but not 1000 ng/ml. CONCLUSIONS OPN deficiency accelerated the spontaneous development of colitis in mice with disrupted gut microbiota and macrophage phagocytic activity.
Collapse
Affiliation(s)
- Takahiko Toyonaga
- Department of Gastroenterology and Hepatology, Kansai Medical University, Hirakata, Osaka, Japan
| | - Hiroshi Nakase
- Department of Gastroenterology and Hepatology, Graduate School of Medicine, Kyoto University, Sakyo-ku, Kyoto, Japan
- * E-mail:
| | - Satoru Ueno
- Department of Internal medicine, Takashima Municipal Hospital, Takashima, Shiga, Japan
| | - Minoru Matsuura
- Department of Gastroenterology and Hepatology, Graduate School of Medicine, Kyoto University, Sakyo-ku, Kyoto, Japan
| | - Takuya Yoshino
- Department of Gastroenterology and Hepatology, Graduate School of Medicine, Kyoto University, Sakyo-ku, Kyoto, Japan
| | - Yusuke Honzawa
- Department of Gastroenterology and Hepatology, Graduate School of Medicine, Kyoto University, Sakyo-ku, Kyoto, Japan
| | - Ayako Itou
- Nutritional Science Institute, Morinaga Milk Industry Co., Ltd, Zama, Kanagawa, Japan
| | - Kazuyoshi Namba
- Nutritional Science Institute, Morinaga Milk Industry Co., Ltd, Zama, Kanagawa, Japan
| | - Naoki Minami
- Department of Gastroenterology and Hepatology, Graduate School of Medicine, Kyoto University, Sakyo-ku, Kyoto, Japan
| | - Satoshi Yamada
- Department of Gastroenterology and Hepatology, Graduate School of Medicine, Kyoto University, Sakyo-ku, Kyoto, Japan
| | - Yorimitsu Koshikawa
- Department of Gastroenterology and Hepatology, Graduate School of Medicine, Kyoto University, Sakyo-ku, Kyoto, Japan
| | - Toshimitsu Uede
- Department of Matrix Medicine, Institute for Genetic Medicine, Hokkaido University, Sapporo, Hokkaido, Japan
| | - Tsutomu Chiba
- Department of Gastroenterology and Hepatology, Graduate School of Medicine, Kyoto University, Sakyo-ku, Kyoto, Japan
| | - Kazuichi Okazaki
- Department of Gastroenterology and Hepatology, Kansai Medical University, Hirakata, Osaka, Japan
| |
Collapse
|
33
|
Osteopontin knockout does not influence the severity of rectal damage in a preclinical model of radiation proctitis in mice. Dig Dis Sci 2015; 60:1633-44. [PMID: 25577272 DOI: 10.1007/s10620-014-3520-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/28/2014] [Accepted: 12/30/2014] [Indexed: 12/13/2022]
Abstract
BACKGROUND Radiation damage to the normal gut is a dose-limiting factor in the application of radiation therapy to treat abdominal and pelvic cancers. All tissue cell types react in concert to orchestrate an acute inflammatory reaction followed by a delayed chronic scarring process. Osteopontin (OPN) is a matricellular protein known to be involved in various physiological but also pathological processes such as tissue inflammation and fibrosis. AIMS The aim of our study was to determine whether OPN knockout influences the severity of radiation proctitis and to investigate the role of OPN in the development of radiation-induced gut damage. RESULTS Here we show that human radiation proctitis is associated with increased immunostaining of the intracellular and extracellular/matrix-linked isoforms of OPN. Moreover, endothelial cells in vitro and rectal tissue in a preclinical model of radiation proctitis in mice both respond to radiation exposure by a sustained increase in OPN mRNA and protein levels. Genetic deficiency of OPN did not influence radiation-induced rectal damage and was associated with significantly decreased animal survival. The acute and late radiation injury scores were similar in OPN-null mice compared with their control littermates. CONCLUSION This study shows that in our model and given the pleiotropic actions of OPN in tissue inflammation and fibrosis, further studies are necessary to understand the precise roles of OPN in radiation-induced proctitis and to determine whether OPN is a useful therapeutic tool in prevention of radiation-induced intestinal tissue injury.
Collapse
|
34
|
Li F, Ye D, Duan J, Sun H. Osteopontin can decrease the cartilage cellular inflammatory reaction induced by LPS. Int J Clin Exp Med 2015; 8:3889-3894. [PMID: 26064289 PMCID: PMC4443123] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2014] [Accepted: 02/06/2015] [Indexed: 06/04/2023]
Abstract
OBJECTIVE This study aims to explore the effect of osteopontin on the inflammatory reaction of cartilage cells stimulated by LPS and its possible signal pathway. METHODS The inflammatory reaction of cartilage cells was induced by LPS, and then the cells were treated with OPN and PD98059 respectively. The expression of TNF-α, IL-1β and ERK1/2 were detected by ELISA or Western-blotting methods. RESULTS Osteopontin could decrease the cartilage cellular inflammatory reaction induced by LPS with dose dependent, while PD98059 could reverse the inhibition of osteopontin. CONCLUSION Osteopontin could decrease the cartilage cellular inflammatory reaction induced by LPS, which may be associated with the ERK1/2 signal pathway.
Collapse
|
35
|
Chai Y, Wang J, Gao Y, Wang T, Shi F, Su J, Yang Y, Zhou X, Song L, Liu Z. Identification of biomarkers for radiation-induced acute intestinal symptoms (RIAISs) in cervical cancer patients by serum protein profiling. JOURNAL OF RADIATION RESEARCH 2015; 56:134-40. [PMID: 25256248 PMCID: PMC4572598 DOI: 10.1093/jrr/rru081] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/11/2023]
Abstract
Radiation-induced acute intestinal symptoms (RIAISs) are the most frequent complication of radiotherapy that causes great pain and limits the treatment efficacy. The aim of this study was to identify serum biomarkers of RIAISs in cervical cancer patients by surface-enhanced laser desorption/ionization time-of-flight mass spectrometry (SELDI-TOF MS). Serum samples were collected from 66 cervical cancer patients prior to pelvic radiotherapy. In our study, RIAISs occurred in 11 patients. An additional 11 patients without RIAISs were selected as controls, whose age, stage, histological type and treatment methods were matched to RIAISs patients. The 22 sera were subsequently analyzed by SELDI-TOF MS, and the resulting protein profiles were evaluated to identify biomarkers using appropriate bioinformatics tools. Comparing the protein profiles of serum samples from the RIAIS group and the control group, it was found that 22 protein peaks were significantly different (P < 0.05), and six of these peaks with mass-to-charge (m/z) ratios of 7514.9, 4603.94, 6887.41, 2769.21, 3839.72 and 4215.7 were successfully identified. A decision tree model of biomarkers was constructed based on three biomarkers (m/z 1270.88, 1503.23 and 7514.90), which separated RIAIS-affected patients from the control group with an accuracy of 81%. This study suggests that serum proteomic analysis by SELDI-TOF MS can identify cervical cancer patients that are susceptible to RIAISs prior to pelvic radiotherapy.
Collapse
Affiliation(s)
- Yanlan Chai
- Department of Radiotherapy Oncology, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, P. R. China
| | - Juan Wang
- Department of Radiotherapy Oncology, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, P. R. China
| | - Ying Gao
- Department of Radiotherapy Oncology, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, P. R. China
| | - Tao Wang
- Department of Radiotherapy Oncology, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, P. R. China
| | - Fan Shi
- Department of Radiotherapy Oncology, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, P. R. China
| | - Jin Su
- Department of Radiotherapy Oncology, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, P. R. China
| | - Yunyi Yang
- Department of Radiotherapy Oncology, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, P. R. China
| | - Xi Zhou
- Department of Radiotherapy Oncology, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, P. R. China Renmin Hospital, Hubei University of Medicine, Hubei 442000, P. R. China
| | - Liping Song
- Department of Radiotherapy Oncology, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, P. R. China
| | - Zi Liu
- Department of Radiotherapy Oncology, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, P. R. China
| |
Collapse
|
36
|
Broom L, Jenner P, Rose S. Increased neurotrophic factor levels in ventral mesencephalic cultures do not explain the protective effect of osteopontin and the synthetic 15-mer RGD domain against MPP+ toxicity. Exp Neurol 2014; 263:1-7. [PMID: 25218309 DOI: 10.1016/j.expneurol.2014.09.005] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2014] [Revised: 09/02/2014] [Accepted: 09/05/2014] [Indexed: 01/13/2023]
Abstract
The synthetic 15-mer arginine-glycine-aspartic acid (RGD) domain of osteopontin (OPN) is protective in vitro and in vivo against dopaminergic cell death and this protective effect may be mediated through interaction with integrin receptors to regulate neurotrophic factor levels. We now examine this concept in rat primary ventral mesencephalic (VM) cultures. 1-Methyl-4-phenylpyridinium (MPP+) exposure reduced tyrosine hydroxylase (TH)-positive cell number and activated glial cells as shown by increased glial fibrillary acidic protein (GFAP), oxycocin-42 (OX-42) and ectodermal dysplasia 1 (ED-1) immunoreactivity. Both OPN and the RGD domain of OPN were equally protective against MPP+ toxicity in VM cultures and both increased glial-derived neurotrophic factor (GDNF) and brain-derived neurotrophic factor (BDNF) levels. The effects of OPN and the RGD domain were accompanied by a decrease in numbers of activated microglia but with no change in astrocyte number. However, full-length OPN and the RGD domain of OPN remained protective against MPP+ toxicity in the presence of a GDNF neutralising antibody. This suggests that increased GDNF levels do not underlie the protective effect observed with OPN. Rather, OPN's protective effect may be mediated through decreased glial cell activation.
Collapse
Affiliation(s)
- Lauren Broom
- Neurodegenerative Diseases Research Group, Institute of Pharmaceutical Science, School of Biomedical Sciences, King's College London, London, SE1 1UL, UK.
| | - Peter Jenner
- Neurodegenerative Diseases Research Group, Institute of Pharmaceutical Science, School of Biomedical Sciences, King's College London, London, SE1 1UL, UK
| | - Sarah Rose
- Neurodegenerative Diseases Research Group, Institute of Pharmaceutical Science, School of Biomedical Sciences, King's College London, London, SE1 1UL, UK
| |
Collapse
|
37
|
Ng JY, Luk FO, Lai TY, Pang CP. Influence of molecular genetics in Vogt-Koyanagi-Harada disease. J Ophthalmic Inflamm Infect 2014; 4:20. [PMID: 25097674 PMCID: PMC4105881 DOI: 10.1186/s12348-014-0020-1] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2014] [Accepted: 07/08/2014] [Indexed: 01/13/2023] Open
Abstract
Vogt-Koyanagi-Harada (VKH) disease is a systemic autoimmune disorder against melanocytes. Recent studies have identified multiple genetic factors that might be associated with the pathogenesis of VKH disease. We performed an electronic database search of PubMed, MEDLINE, and EMBASE, and all relevant papers published up to 13 June 2014 were reviewed. A total of 1,031 publications including articles relevant to the genetics of VKH disease and the references of these articles were reviewed. The review identified a number of genetic factors which might be involved in the pathogenesis of VKH disease, some of which may alter the clinical course of VKH disease. Genes which might be involved in the pathogenesis of VKH disease included genes expressing HLA, complement factor H, interleukins, cytotoxic T-lymphocyte antigen 4 (CTLA-4), killer cell immunoglobulin-like receptors (KIR), programmed cell death 1 (PDCD1), protein tyrosine phosphatase non-receptor 22 (PTPN22), osteopontin, tumor necrosis factor alpha-induced protein 3 (TNFAIP3), macrophage migration inhibitory factor (MIF), and other immune response genes. Further studies to explore the correlation among different genotypes and phenotypes of VKH disease will be useful to shed light on the pathogenesis of uveitis in VKH disease and may facilitate the development of new treatment modalities of uveitis in VKH disease.
Collapse
Affiliation(s)
- Joanne Yw Ng
- Department of Ophthalmology and Visual Sciences, The Chinese University of Hong Kong, 3/F Hong Kong Eye Hospital, 147K Argyle Street, Kowloon ᅟ, Hong Kong
| | - Fiona Oj Luk
- Department of Ophthalmology and Visual Sciences, The Chinese University of Hong Kong, 3/F Hong Kong Eye Hospital, 147K Argyle Street, Kowloon ᅟ, Hong Kong
| | - Timothy Yy Lai
- Department of Ophthalmology and Visual Sciences, The Chinese University of Hong Kong, 3/F Hong Kong Eye Hospital, 147K Argyle Street, Kowloon ᅟ, Hong Kong
| | - Chi-Pui Pang
- Department of Ophthalmology and Visual Sciences, The Chinese University of Hong Kong, 3/F Hong Kong Eye Hospital, 147K Argyle Street, Kowloon ᅟ, Hong Kong
| |
Collapse
|
38
|
Kaleta B. Role of osteopontin in systemic lupus erythematosus. Arch Immunol Ther Exp (Warsz) 2014; 62:475-82. [PMID: 24917428 PMCID: PMC4244532 DOI: 10.1007/s00005-014-0294-x] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2014] [Accepted: 04/07/2014] [Indexed: 12/23/2022]
Abstract
Systemic lupus erythematosus (SLE) is a multisystemic disease, caused by a variety of factors, which lead to immunological abnormalities. Osteopontin (OPN) is a pleiotropic protein, important in bone remodeling and immune system signaling. OPN, produced by various cells, including immune cells, plays a key role in regulating T-helper 1/T-helper 2 balance, stimulating B lymphocytes to produce antibodies, regulating macrophages, neutrophils and inducing dendritic cells. OPN expression is influenced by genetic polymorphisms of its promoter, hormones and cytokines. Over expression of OPN has been associated with the pathogenesis of immune-mediated diseases. OPN has been implicated in the development of murine model of lupus and in humans with SLE. In this review, I will present current state of research on the role of OPN and OPN gene polymorphisms in pathogenesis and clinical course of SLE. A better understanding of the role of OPN in SLE will contribute to more precise diagnosis and treatment of the disease.
Collapse
Affiliation(s)
- Beata Kaleta
- Department of Clinical Immunology, Transplantation Institute, Medical University of Warsaw, Nowogrodzka 59, 02-006, Warsaw, Poland,
| |
Collapse
|
39
|
Joung KE, Christou H, Park KH, Mantzoros CS. Cord blood levels of osteopontin as a phenotype marker of gestational age and neonatal morbidities. Obesity (Silver Spring) 2014; 22:1317-24. [PMID: 24123979 PMCID: PMC4394598 DOI: 10.1002/oby.20626] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/29/2013] [Accepted: 09/13/2013] [Indexed: 11/07/2022]
Abstract
OBJECTIVES Osteopontin (OPN) is a proinflammatory cytokine associated with metabolic syndrome. Extreme birth weight categories including small for gestational age (SGA), and large for gestational age (LGA) are risk factors for metabolic syndrome. However normal levels of plasma OPN in neonates and the relationship of OPN to fetal growth remain unknown. We evaluated the association of umbilical cord blood OPN with gestational age, birth weight, and neonatal outcomes. METHODS A cross-sectional study of 261 newborns of all gestational ages beginning at week 26, and 26 adults for comparison was performed. Umbilical cord blood from newborns and analyzed plasma for OPN by ELISA was collected. RESULTS Plasma OPN was significantly higher in neonates (414.65 ± 136.72 ng/mL) compared to adults (33.37 ± 14.66 ng/mL, P < 0.001). There was an inverse correlation between OPN and gestational age (r = -0.48, P < 0.0001). LGA infants had lower OPN than appropriate for gestational age (AGA) infants, but LGA was not an independent predictor of OPN in multivariate analysis. Among preterm infants, patent ductus arteriosus (PDA) was independently associated with higher OPN (OR = 2.49, P = 0.02). CONCLUSION Our results raise the possibility that OPN has a physiologic role in fetal growth and development, and may be a useful biomarker for PDA.
Collapse
Affiliation(s)
- Kyoung Eun Joung
- Division of Newborn Medicine, Boston Children's Hospital, Enders-961 300 Longwood Ave, Boston, MA, 02115; Division of Newborn Medicine, Brigham and Women's Hospital (BWH), 75 Francis St, Boston, MA, 02115
| | | | | | | |
Collapse
|
40
|
Ge X, Leung TM, Arriazu E, Lu Y, Urtasun R, Christensen B, Fiel MI, Mochida S, Sørensen ES, Nieto N. Osteopontin binding to lipopolysaccharide lowers tumor necrosis factor-α and prevents early alcohol-induced liver injury in mice. Hepatology 2014; 59:1600-16. [PMID: 24214181 PMCID: PMC3966944 DOI: 10.1002/hep.26931] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/15/2013] [Accepted: 11/06/2013] [Indexed: 12/16/2022]
Abstract
UNLABELLED Although osteopontin (OPN) is induced in alcoholic patients, its role in the pathophysiology of alcoholic liver disease (ALD) remains unclear. Increased translocation of lipopolysaccharide (LPS) from the gut is key for the onset of ALD because it promotes macrophage infiltration and activation, tumor necrosis factor-α (TNFα) production, and liver injury. Since OPN is protective for the intestinal mucosa, we postulated that enhancing OPN expression in the liver and consequently in the blood and/or in the gut could protect from early alcohol-induced liver injury. Wild-type (WT), OPN knockout (Opn(-/-)), and transgenic mice overexpressing OPN in hepatocytes (Opn(HEP) Tg) were fed either the control or the ethanol Lieber-DeCarli diet. Ethanol increased hepatic, plasma, biliary, and fecal OPN more in Opn(HEP) Tg than in WT mice. Steatosis was less in ethanol-treated Opn(HEP) Tg mice as shown by decreased liver-to-body weight ratio, hepatic triglycerides, the steatosis score, oil red-O staining, and lipid peroxidation. There was also less inflammation and liver injury as demonstrated by lower alanine aminotransferase (ALT) activity, hepatocyte ballooning degeneration, LPS levels, the inflammation score, and the number of macrophages and TNFα(+) cells. To establish if OPN could limit LPS availability and its noxious effects in the liver, binding studies were performed. OPN showed binding affinity for LPS which prevented macrophage activation, reactive oxygen, and nitrogen species generation and TNFα production. Treatment with milk OPN (m-OPN) blocked LPS translocation in vivo and protected from early alcohol-induced liver injury. CONCLUSION Natural induction plus forced overexpression of OPN in the liver or treatment with m-OPN protect from early alcohol-induced liver injury by blocking the gut-derived LPS and TNFα effects in the liver.
Collapse
Affiliation(s)
- Xiaodong Ge
- Division of Liver Diseases, Department of Medicine, Mount Sinai School of Medicine, Box 1123, 1425 Madison Avenue, Room 11-76, New York, NY 10029, USA
| | - Tung-Ming Leung
- Division of Liver Diseases, Department of Medicine, Mount Sinai School of Medicine, Box 1123, 1425 Madison Avenue, Room 11-76, New York, NY 10029, USA
| | - Elena Arriazu
- Division of Liver Diseases, Department of Medicine, Mount Sinai School of Medicine, Box 1123, 1425 Madison Avenue, Room 11-76, New York, NY 10029, USA
| | - Yongke Lu
- Division of Liver Diseases, Department of Medicine, Mount Sinai School of Medicine, Box 1123, 1425 Madison Avenue, Room 11-76, New York, NY 10029, USA
| | - Raquel Urtasun
- Division of Liver Diseases, Department of Medicine, Mount Sinai School of Medicine, Box 1123, 1425 Madison Avenue, Room 11-76, New York, NY 10029, USA
| | - Brian Christensen
- Department of Molecular Biology and Genetics, Aarhus University, Gustav Wieds Vej 10, Aarhus Science Park, DK-8000 Denmark
| | - Maria Isabel Fiel
- Department of Pathology, Mount Sinai School of Medicine, 1468 Madison Avenue, Room 15-28A, New York, NY 10029, USA
| | - Satoshi Mochida
- Gastroenterology and Hepatology, Internal Medicine, Saitama Medical School, 38 Morohongo, Moroyama-cho, Iruma-gun, Saitama 350-0495, Japan
| | - Esben S. Sørensen
- Department of Molecular Biology and Genetics, Aarhus University, Gustav Wieds Vej 10, Aarhus Science Park, DK-8000 Denmark
| | - Natalia Nieto
- Division of Liver Diseases, Department of Medicine, Mount Sinai School of Medicine, Box 1123, 1425 Madison Avenue, Room 11-76, New York, NY 10029, USA
| |
Collapse
|
41
|
Osteopontin expression by CD103- dendritic cells drives intestinal inflammation. Proc Natl Acad Sci U S A 2014; 111:E856-65. [PMID: 24550510 DOI: 10.1073/pnas.1316447111] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Intestinal CD103(-) dendritic cells (DCs) are pathogenic for colitis. Unveiling molecular mechanisms that render these cells proinflammatory is important for the design of specific immunotherapies. In this report, we demonstrated that mesenteric lymph node CD103(-) DCs express, among other proinflammatory cytokines, high levels of osteopontin (Opn) during experimental colitis. Opn expression by CD103(-) DCs was crucial for their immune profile and pathogenicity, including induction of T helper (Th) 1 and Th17 cell responses. Adoptive transfer of Opn-deficient CD103(-) DCs resulted in attenuated colitis in comparison to transfer of WT CD103(-) DCs, whereas transgenic CD103(-) DCs that overexpress Opn were highly pathogenic in vivo. Neutralization of secreted Opn expressed exclusively by CD103(-) DCs restrained disease severity. Also, Opn deficiency resulted in milder disease, whereas systemic neutralization of secreted Opn was therapeutic. We determined a specific domain of the Opn protein responsible for its CD103(-) DC-mediated proinflammatory effect. We demonstrated that disrupting the interaction of this Opn domain with integrin α9, overexpressed on colitic CD103(-) DCs, suppressed the inflammatory potential of these cells in vitro and in vivo. These results add unique insight into the biology of CD103(-) DCs and their function during inflammatory bowel disease.
Collapse
|
42
|
Increased osteopontin expression in HTLV-1-associated myelopathy/tropical spastic paraparesis (HAM/TSP) patient cells is associated with IL-17 expression. J Clin Virol 2013; 58:295-8. [PMID: 23756056 DOI: 10.1016/j.jcv.2013.05.006] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2012] [Revised: 04/12/2013] [Accepted: 05/06/2013] [Indexed: 01/18/2023]
Abstract
BACKGROUND HTLV-1-associated myelopathy/tropical spastic paraparesis (HAM/TSP) is a neurological inflammatory disease associated with a predominant infiltration of CD4+ T lymphocytes, which are the main subset of HTLV-1-infected cells. It has been demonstrated that in cell line the viral Tax protein transcriptionnally regulate expression of osteopontin, an inflammatory cytokine associated with Th17-related pathologies. OBJECTIVES The aim of the study was to explore osteopontin expression in HTLV-1 asymptomatic carriers and in HAM/TSP patients and consequences on IL17 expression. STUDY DESIGN We quantified Tax, osteopontin, RORγ, IL17 and IL22 mRNA expressions in cells from 10 HAM/TSP patients, 6 asymptomatic HTLV-1 carriers (ASY) and 4 HTLV-1-negative healthy donors during ex vivo culture. RESULTS We observed that the expression of osteopontin was higher in HAM/TSP patients and correlated with Tax expression levels. Positive regulation of RORγ, IL17 and IL22 were also observed during cell culture. CONCLUSIONS Our results propose a new mechanism which could contribute to HAM/TSP pathogenesis.
Collapse
|
43
|
Osteopontin: participation in inflammation or mucosal protection in inflammatory bowel diseases? Dig Dis Sci 2013; 58:1569-80. [PMID: 23361573 DOI: 10.1007/s10620-012-2556-y] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/15/2012] [Accepted: 12/24/2012] [Indexed: 12/22/2022]
Abstract
BACKGROUND Osteopontin (OPN) is associated with the Th1 immune response in inflammatory bowel diseases (IBD). While OPN has been shown to play an important role in maintaining the epithelial barrier, its role in IBD remains unclear. AIM The aim of this study was to assess OPN function in patients with IBD and in the mouse colitis model. METHODS Osteopontin expression in colonic samples from IBD patients was determined by a semi-quantitative immunohistochemical staining method. Colitis in BALB/c mice was induced by 5 % dextran sodium sulfate (DSS), followed by treatment with salazosulfapyridine (SASP) and infliximab, respectively. The plasma OPN concentration was measured by an enzyme-linked immunosorbent assay. The expression of OPN in colonic tissues was detected by reverse transcriptase PCR, real-time PCR and Western blot, and the localization of OPN was determined by a semi-quantitative immunohistochemical staining method. The immune function of OPN was investigated by measuring the production of cytokines, and the amount of cytokines produced was then used to determine OPN immune functions. RESULTS Osteopontin expression in intestinal epithelial cells was significantly lower in IBD patients than in controls, while its expression in lamina propria exudative cells was significantly higher in IBD patients than in controls. In DSS-induced mice, OPN expression in plasma and colonic tissues increased significantly, and this increase was significantly reduced after the mice were treated with SASP and infliximab. OPN promoted the Th1 immune response and strengthened inflammation in the mouse colitis model. CONCLUSIONS Our results indicate that OPN plays an important role in the immune response and is also involved in the mucosal protective mechanism in IBD.
Collapse
|
44
|
Jiang Y, Yao M, Liu Q, Zhou C. OPN gene polymorphisms influence the risk of knee OA and OPN levels in synovial fluid in a Chinese population. Arthritis Res Ther 2013; 15:R3. [PMID: 23289928 PMCID: PMC3672660 DOI: 10.1186/ar4129] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2012] [Accepted: 12/20/2012] [Indexed: 12/17/2022] Open
Abstract
INTRODUCTION A body of studies suggests the role of osteopontin (OPN) in onset and development of osteoarthritis (OA), however, the association between OPN polymorphisms and OA susceptibility as well as its clinical features has not been reported. METHODS A total of 750 patients with primary knee OA and 794 healthy volunteer were enrolled as controls. Both OA and control groups were interviewed to obtain demographic and clinical data. Three polymorphisms of OPN gene, namely, -156GG/G, -443C/T and -66T/G were determined. The levels of the full length and the thrombin-cleaved OPN in synovial fluid (SF) from OA subjects were measured. RESULTS We found the polymorphisms of the -443C/T and the -66/T/G were significantly associated with the OA risk and the radiographic severity. The -443TT and -66GG showed protective effect against developing OA and were associated with lower Kellgren-Lawrence grade. Besides, the polymorphisms of -443C/T and -66T/G significantly affected the thrombin-cleaved OPN levels in SF from OA subjects. Subjects with -443TT and -66GG genotypes had lower thrombin-cleaved OPN levels in SF. The thrombin-cleaved OPN levels in SF were positively correlated to the radiographic severity of OA. CONCLUSIONS Our findings suggest that certain OPN gene polymorphisms may be used as molecular markers for the susceptibility and severity of OA.
Collapse
|
45
|
Shin T. Osteopontin as a two-sided mediator in acute neuroinflammation in rat models. Acta Histochem 2012; 114:749-54. [PMID: 22947282 DOI: 10.1016/j.acthis.2012.08.004] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2012] [Revised: 08/13/2012] [Accepted: 08/14/2012] [Indexed: 01/26/2023]
Abstract
Osteopontin (OPN) plays an important role in the initiation of inflammation, affecting cell adhesion, chemotaxis, immune regulation, and protection against apoptosis, depending on its intracellular or extracellular localization. Although OPN in inflammation of the autoimmune central nervous system is proinflammatory, recent studies have shown that OPN during the induction stage of inflammation may also participate in neuroprotection and neurite growth. The present review examines the dual roles of OPN, specifically, its proinflammatory and subsequent neuroprotective roles, in acute neuroinflammation in rat models, including experimental autoimmune encephalomyelitis, brain injury, and autoimmune neuritis. All of these models are characterized by acute neuroinflammation, followed by remodeling of neural tissues.
Collapse
MESH Headings
- Acute Disease
- Animals
- Brain Injuries/immunology
- Brain Injuries/metabolism
- Brain Injuries/pathology
- Disease Models, Animal
- Encephalomyelitis, Autoimmune, Experimental/immunology
- Encephalomyelitis, Autoimmune, Experimental/metabolism
- Encephalomyelitis, Autoimmune, Experimental/pathology
- Inflammation
- Inflammation Mediators/metabolism
- Neuritis, Autoimmune, Experimental/immunology
- Neuritis, Autoimmune, Experimental/metabolism
- Neuritis, Autoimmune, Experimental/pathology
- Osteopontin/metabolism
- Rats
Collapse
Affiliation(s)
- Taekyun Shin
- Department of Veterinary Anatomy, College of Veterinary Medicine, Jeju National University, Republic of Korea.
| |
Collapse
|
46
|
Shaker OG, Sadik NA, El-Dessouki A. Single-nucleotide polymorphism in the promoter region of the osteopontin gene at nucleotide −443 as a marker predicting the efficacy of pegylated interferon/ribavirin-therapy in Egyptians patients with chronic hepatitis C. Hum Immunol 2012; 73:1039-45. [DOI: 10.1016/j.humimm.2012.07.329] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2012] [Revised: 05/02/2012] [Accepted: 07/13/2012] [Indexed: 10/28/2022]
|
47
|
Oz HS, Zhong J, de Villiers WJS. Osteopontin ablation attenuates progression of colitis in TNBS model. Dig Dis Sci 2012; 57:1554-61. [PMID: 22173746 DOI: 10.1007/s10620-011-2009-z] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/20/2011] [Accepted: 11/29/2011] [Indexed: 12/20/2022]
Abstract
INTRODUCTION OPN has been implicated in the inflammatory response to Crohn's disease. We hypothesized that OPN deficiency protects against different stages of TNBS-induced colitis in a modified model that mimics Crohn's disease. MATERIAL AND METHODS OPN-deficient and wildtype mice were treated intracolonically with TNBS and euthanized during acute, sub-acute and chronic colitis. RESULTS TNBS-treated wildtype mice developed severe colitis, but OPN-deficient mice were significantly protected. Wildtype mice showed significant infiltration of inflammatory cells including macrophages, and colonic transmural thickening that progressed to strictures, increased matrix collagen deposits (X2 fold), and granuloma formation. These pathological findings were partially attenuated by OPN deficiency. The inflammatory marker, serum amyloid A (SAA), markedly increased in sub-acute stages regardless of OPN status. Conversely, OPN deficiency significantly reduced concentration of SAA in the acute and chronic stages. Secretory OPN was upregulated particularly in acute stage in wildtypes (P < 0.001) and as expected not present in OPN-deficient animals. Flow cytometry analysis of splenic macrophages revealed significant increases in scavenger receptors, macrosialin and F4/80 markers' expression in wildtypes. CONCLUSIONS Our data support the role of OPN in induction of inflammation and establishment of chronic colitis. Therefore, OPN may represent a target for therapeutic intervention in Crohn's disease.
Collapse
Affiliation(s)
- Helieh S Oz
- Department of Physiology, University of Kentucky Medical Center, Lexington, KY 40515-0298, USA.
| | | | | |
Collapse
|
48
|
Glas J, Seiderer J, Bayrle C, Wetzke M, Fries C, Tillack C, Olszak T, Beigel F, Steib C, Friedrich M, Diegelmann J, Czamara D, Brand S. The role of osteopontin (OPN/SPP1) haplotypes in the susceptibility to Crohn's disease. PLoS One 2011; 6:e29309. [PMID: 22242114 PMCID: PMC3248444 DOI: 10.1371/journal.pone.0029309] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2011] [Accepted: 11/25/2011] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND Osteopontin represents a multifunctional molecule playing a pivotal role in chronic inflammatory and autoimmune diseases. Its expression is increased in inflammatory bowel disease (IBD). The aim of our study was to analyze the association of osteopontin (OPN/SPP1) gene variants in a large cohort of IBD patients. METHODOLOGY/PRINCIPAL FINDINGS Genomic DNA from 2819 Caucasian individuals (n = 841 patients with Crohn's disease (CD), n = 473 patients with ulcerative colitis (UC), and n = 1505 healthy unrelated controls) was analyzed for nine OPN SNPs (rs2728127, rs2853744, rs11730582, rs11739060, rs28357094, rs4754 = p.Asp80Asp, rs1126616 = p.Ala236Ala, rs1126772 and rs9138). Considering the important role of osteopontin in Th17-mediated diseases, we performed analysis for epistasis with IBD-associated IL23R variants and analyzed serum levels of the Th17 cytokine IL-22. For four OPN SNPs (rs4754, rs1126616, rs1126772 and rs9138), we observed significantly different distributions between male and female CD patients. rs4754 was protective in male CD patients (p = 0.0004, OR = 0.69). None of the other investigated OPN SNPs was associated with CD or UC susceptibility. However, several OPN haplotypes showed significant associations with CD susceptibility. The strongest association was found for a haplotype consisting of the 8 OPN SNPs rs2728127-rs2853744-rs11730582-rs11439060-rs28357094-rs112661-rs1126772-rs9138 (omnibus p-value = 2.07×10⁻⁸). Overall, the mean IL-22 secretion in the combined group of OPN minor allele carriers with CD was significantly lower than that of CD patients with OPN wildtype alleles (p = 3.66×10⁻⁵). There was evidence for weak epistasis between the OPN SNP rs28357094 with the IL23R SNP rs10489629 (p = 4.18×10⁻²) and between OPN SNP rs1126616 and IL23R SNP rs2201841 (p = 4.18×10⁻²) but none of these associations remained significant after Bonferroni correction. CONCLUSIONS/SIGNIFICANCE Our study identified OPN haplotypes as modifiers of CD susceptibility, while the combined effects of certain OPN variants may modulate IL-22 secretion.
Collapse
Affiliation(s)
- Jürgen Glas
- Department of Medicine II - Grosshadern, Ludwig-Maximilians-University, Munich, Germany
- Department of Preventive Dentistry and Periodontology, Ludwig-Maximilians-University, Munich, Germany
- Department of Human Genetics, Rheinisch-Westfälische Technische Hochschule (RWTH), Aachen, Germany
| | - Julia Seiderer
- Department of Medicine II - Grosshadern, Ludwig-Maximilians-University, Munich, Germany
| | - Corinna Bayrle
- Department of Preventive Dentistry and Periodontology, Ludwig-Maximilians-University, Munich, Germany
| | - Martin Wetzke
- Department of Pediatrics, Hannover Medical School, Hannover, Germany
| | - Christoph Fries
- Department of Medicine II - Grosshadern, Ludwig-Maximilians-University, Munich, Germany
- Department of Preventive Dentistry and Periodontology, Ludwig-Maximilians-University, Munich, Germany
| | - Cornelia Tillack
- Department of Medicine II - Grosshadern, Ludwig-Maximilians-University, Munich, Germany
| | - Torsten Olszak
- Department of Medicine II - Grosshadern, Ludwig-Maximilians-University, Munich, Germany
- Division of Gastroenterology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Florian Beigel
- Department of Medicine II - Grosshadern, Ludwig-Maximilians-University, Munich, Germany
| | - Christian Steib
- Department of Medicine II - Grosshadern, Ludwig-Maximilians-University, Munich, Germany
| | - Matthias Friedrich
- Department of Medicine II - Grosshadern, Ludwig-Maximilians-University, Munich, Germany
- Department of Preventive Dentistry and Periodontology, Ludwig-Maximilians-University, Munich, Germany
| | - Julia Diegelmann
- Department of Medicine II - Grosshadern, Ludwig-Maximilians-University, Munich, Germany
- Department of Preventive Dentistry and Periodontology, Ludwig-Maximilians-University, Munich, Germany
| | | | - Stephan Brand
- Department of Medicine II - Grosshadern, Ludwig-Maximilians-University, Munich, Germany
| |
Collapse
|
49
|
Hsieh YH, Margaret Juliana M, Ho KJ, Kuo HC, van der Heyde H, Elmets C, Chang PL. Host-derived osteopontin maintains an acute inflammatory response to suppress early progression of extrinsic cancer cells. Int J Cancer 2011; 131:322-33. [PMID: 21826648 DOI: 10.1002/ijc.26359] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2010] [Accepted: 07/27/2011] [Indexed: 01/13/2023]
Abstract
The matricellular protein osteopontin (OPN), expressed in various cancer types and elevated in the blood of cancer patients, is thought to have different functions when derived from host versus cancer cells. To assess the effect of host-derived OPN on growth of cancers of epithelial origin, we established a line of cutaneous squamous cell carcinoma (SCC) cells, named ONSC, which lacks the OPN gene and develops SCC in syngeneic wild-type (WT) and OPN-null mice. At 8 and/or 10 week after subcutaneous injection of ONSC cells in mice, however, there was a lower tumor incidence in WT mice, suggesting that host-derived OPN is associated with suppression of early growth of extrinsic cancer cells. Histological, immunohistochemical, biochemical and hematological analyses were performed on the tumor microenvironment and blood from tumor-bearing mice during the first week after implantation. Host-derived OPN suppression of extrinsic ONSC cell progression is likely mediated through elicitation of an early innate inflammatory response, through its function as a chemoattractant and/or by enhancing survival of inflammatory cells. Further, consistent with a previous report, the serum levels of host-derived OPN, which are elevated during the early phase of tumor growth in mice implanted with ONSC, appear to reflect an anti-tumor progression effect.
Collapse
Affiliation(s)
- Yu-Hua Hsieh
- Department of Nutrition Sciences, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | | | | | | | | | | | | |
Collapse
|
50
|
Abstract
Osteopontin (OPN) is a protein involved in various pathophysiological events. OPN has been studied as a secreted protein, but recent reports showed that OPN can be found in the cytoplasm and the nucleus. Therefore, some OPN molecules are not secreted and stay in cells. Such intracellular OPN (iOPN) has biological functions distinct from secreted OPN (sOPN). iOPN is involved in cytoskeletal rearrangement and in signal transduction pathways downstream of innate immune receptors, such as Toll-like receptors (TLRs), as an adaptor or scaffolding protein. Although sOPN and iOPN are generated from the same Opn mRNA species, biological outcomes mediated by two isoforms can be different. It would be necessary to delineate which isoform of OPN is responsible for pathophysiological events.
Collapse
|