1
|
Liu C, Xia S, Wang B, Li J, Wang X, Ren Y, Zhou X. Osteopontin promotes tumor microenvironment remodeling and therapy resistance. Cancer Lett 2025; 617:217618. [PMID: 40058726 DOI: 10.1016/j.canlet.2025.217618] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2025] [Revised: 03/05/2025] [Accepted: 03/07/2025] [Indexed: 03/15/2025]
Abstract
Osteopontin (OPN) is a multifunctional secretory protein which can be expressed and secreted by a variety of tumor cells and immune cells. Tumor microenvironment remodeling provides favorable conditions for tumor progression, immune escape and therapy resistance. As a bridge molecule in crosstalk between tumor cells and tumor microenvironment, OPN can not only come from tumor cells to regulate the functions of various immune cells, promoting the formation of immunosuppressive environment, but also can be secreted by immune cells to act on tumor cells, leading to tumor progression, thus constructing a positive feedback regulatory network. Here, we summarize the molecular structure, source and receptor of OPN, and clarify the mechanism of OPN on tumor-associated macrophages, dendritic cells, myeloid-derived suppressor cells, tumor progression and therapy resistance to comprehensively understand the great potential of OPN as a tumor biomarker and therapeutic target.
Collapse
Affiliation(s)
- Chao Liu
- Department of Maxillofacial and Otorhinolaryngological Oncology, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin, 300060, China; Key Laboratory of Basic and Translational Medicine on Head & Neck Cancer, Tianjin, 300060, China; State Key Laboratory of Druggability Evaluation and Systematic Translational Medicine, Tianjin, 300060, China
| | - Shunjin Xia
- Department of Maxillofacial and Otorhinolaryngological Oncology, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin, 300060, China; Key Laboratory of Basic and Translational Medicine on Head & Neck Cancer, Tianjin, 300060, China; State Key Laboratory of Druggability Evaluation and Systematic Translational Medicine, Tianjin, 300060, China
| | - Bo Wang
- Department of Maxillofacial and Otorhinolaryngological Oncology, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin, 300060, China; Key Laboratory of Basic and Translational Medicine on Head & Neck Cancer, Tianjin, 300060, China; State Key Laboratory of Druggability Evaluation and Systematic Translational Medicine, Tianjin, 300060, China
| | - Jiayong Li
- Department of Maxillofacial and Otorhinolaryngological Oncology, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin, 300060, China; Key Laboratory of Basic and Translational Medicine on Head & Neck Cancer, Tianjin, 300060, China; State Key Laboratory of Druggability Evaluation and Systematic Translational Medicine, Tianjin, 300060, China
| | - Xuyan Wang
- Department of Maxillofacial and Otorhinolaryngological Oncology, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin, 300060, China; Key Laboratory of Basic and Translational Medicine on Head & Neck Cancer, Tianjin, 300060, China; State Key Laboratory of Druggability Evaluation and Systematic Translational Medicine, Tianjin, 300060, China
| | - Yu Ren
- Key Laboratory of Basic and Translational Medicine on Head & Neck Cancer, Tianjin, 300060, China; State Key Laboratory of Druggability Evaluation and Systematic Translational Medicine, Tianjin, 300060, China; Department of Genetics, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, 300070, China.
| | - Xuan Zhou
- Department of Maxillofacial and Otorhinolaryngological Oncology, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin, 300060, China; Key Laboratory of Basic and Translational Medicine on Head & Neck Cancer, Tianjin, 300060, China; State Key Laboratory of Druggability Evaluation and Systematic Translational Medicine, Tianjin, 300060, China.
| |
Collapse
|
2
|
Huang Z, Li Y, Liu Q, Chen X, Lin W, Wu W, Chen Z, Chen X, Pan Y, Qiu S. SPP1-mediated M2 macrophage polarization shapes the tumor microenvironment and enhances prognosis and immunotherapy guidance in nasopharyngeal carcinoma. Int Immunopharmacol 2025; 147:113944. [PMID: 39742726 DOI: 10.1016/j.intimp.2024.113944] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Revised: 12/21/2024] [Accepted: 12/22/2024] [Indexed: 01/04/2025]
Abstract
Secreted phosphoprotein 1 (SPP1) shows carcinogenic potential in multiple cancers, yet its role in nasopharyngeal carcinoma (NPC) remains elusive. Leveraging transcriptomic data sourced from an NPC cohort at Fujian Cancer Hospital, alongside datasets from the Gene Expression Omnibus cohort and a single-cell RNA sequencing dataset, this investigation explored the role of SPP1 in tumor progression and the tumor microenvironment of NPC. A co-culture system involving tumor cells and macrophages was established to elucidate the relationship between SPP1 and tumor-associated macrophages in NPC. Subsequently, we established an SPP1-driven M2 macrophage signature using a machine-learning-based framework to predict patient prognosis. The results of our analysis indicated that SPP1 is associated with an elevated risk of disease progression and poor prognosis in NPC. Single-cell analysis demonstrated that SPP1 is a pivotal gene in the polarization of M2 macrophages within the tumor microenvironment. In vitro experiments demonstrated that NPC-derived SPP1 has the potential to activate the CD44/JAK2/STAT3 signaling pathway, promoting macrophage recruitment and polarization of the M2 subtype. Furthermore, we established a comprehensive SPP1-related M2 macrophage signature that can predict the prognosis and immune characteristics of patients with NPC. Our findings offer new insights into the role of SPP1 in the tumor microenvironment of NPC, and provide a novel SPP1-driven M2 macrophage signature with the potential to serve as a valuable tool for prognosis prediction and personalized therapy in NPC.
Collapse
Affiliation(s)
- Zongwei Huang
- Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital (Fujian Branch of Fudan University Shanghai Cancer Center), Fuzhou, China
| | - Ying Li
- Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital (Fujian Branch of Fudan University Shanghai Cancer Center), Fuzhou, China
| | - Qinying Liu
- Fujian Provincial Key Laboratory of Tumor Biotherapy, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, China
| | - Xiaochuan Chen
- Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital (Fujian Branch of Fudan University Shanghai Cancer Center), Fuzhou, China
| | - Wanzun Lin
- Shanghai Proton and Heavy Ion Center, Fudan University Cancer Hospital, Shanghai, China
| | - Wenxi Wu
- Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital (Fujian Branch of Fudan University Shanghai Cancer Center), Fuzhou, China
| | - Zihan Chen
- Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital (Fujian Branch of Fudan University Shanghai Cancer Center), Fuzhou, China
| | - Xin Chen
- Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital (Fujian Branch of Fudan University Shanghai Cancer Center), Fuzhou, China
| | - Yuhui Pan
- Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital (Fujian Branch of Fudan University Shanghai Cancer Center), Fuzhou, China.
| | - Sufang Qiu
- Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital (Fujian Branch of Fudan University Shanghai Cancer Center), Fuzhou, China.
| |
Collapse
|
3
|
Zheng F, Dong T, Chen Y, Wang L, Peng G. Border-associated macrophages: From physiology to therapeutic targets in Alzheimer's disease. Exp Neurol 2025; 383:115021. [PMID: 39461707 DOI: 10.1016/j.expneurol.2024.115021] [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: 06/27/2024] [Revised: 10/15/2024] [Accepted: 10/20/2024] [Indexed: 10/29/2024]
Abstract
Border-associated macrophages (BAMs) constitute a highly heterogeneous group of central nervous system-resident macrophages at the brain boundaries. Despite their significance, BAMs have mainly been overlooked compared to microglia, resulting in a limited understanding of their functions. However, recent advancements in single-cell immunophenotyping and transcriptomic analyses of BAMs have revealed a previously unrecognized complexity in these cells, in addition to their critical roles under non-pathological conditions and diseases like Alzheimer's disease (AD), Parkinson's disease, glioma, and ischemic stroke. In this review, we discuss the origins, self-renewal capabilities, and extensive heterogeneity of BAMs, and clarify their important physiological functions such as immune monitoring, waste removal and vascular permeability regulation. We also summarize experimental evidence linking BAMs to the progression of AD. Finally, we review therapeutic strategies targeting brain innate immune cells mainly focusing on strategies aimed at modulating BAMs to treat AD and evaluate their potential in clinical applications.
Collapse
Affiliation(s)
- Fangxue Zheng
- Department of Neurology, The First Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Taiwei Dong
- Hangzhou Normal University School of Basic Medical Sciences, Hangzhou, China
| | - Yi Chen
- Department of Neurology, The First Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Lang Wang
- Department of Neurology, The First Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China; Interdisciplinary Institute of Neuroscience and Technology, Zhejiang University School of Medicine, Hangzhou, China.
| | - Guoping Peng
- Department of Neurology, The First Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China.
| |
Collapse
|
4
|
Liu C, Ge Y. Immune-Related Genes Associated with Interstitial Lung Disease in Dermatomyositis. Int J Gen Med 2024; 17:5261-5271. [PMID: 39563787 PMCID: PMC11573690 DOI: 10.2147/ijgm.s490294] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2024] [Accepted: 11/09/2024] [Indexed: 11/21/2024] Open
Abstract
Background Interstitial lung disease (ILD) is one of the significant complications of dermatomyositis (DM), but the mechanisms by which it occurs remain incompletely elucidated. This study aimed to explore further the possible genetic mechanisms by which this complication occurs. Methods Gene expression profiles for DM (GSE39454, GSE46239, GSE143323) and ILD (GSE32537, GSE110147, GSE150910) were downloaded from the Gene Expression Omnibus (GEO) database. After identifying common differentially expressed genes (DEGs) to DM and ILD using the "limma" R package and the "VennDiagram" R package, functional annotation, relationship to immune cell infiltration, identification of transcription factors (TFs), we also collected clinical cases of DM-associated ILD (DM-ILD), including 3 cases of rapidly progressive ILD (RP-ILD) and 3 cases of none-RP-ILD, and explored whether there were differences in serum lymphocyte subpopulations. Results A total of 4 common DEGs (SLAMF7, SPP1, TDO2, and VCAM1) were screened and Gene Ontology (GO) enrichment analysis showed that these genes were mainly enriched in T cell activation, regulation of lymphocyte activation, lymphocyte differentiation, leukocyte proliferation and regulation of T cell activation. In terms of Kyoto Encyclopedia of Genes and Genomes (KEGG) pathways, the three significantly enriched pathways were the PI3K-Akt signaling pathway, MAPK signaling pathway, and Cytokine-cytokine receptor interaction. In lung and muscle tissues, 21 and 3 TFs may regulate the expression of these genes, respectively. Finally, by analysing the serum lymphocyte subpopulations, we also found a decrease in the absolute number of CD8+ T cells and an increase in the CD4+ /CD8+ T cell ratio in DM combined with RP-ILD. Conclusion These common pathways and key genes may provide new ideas for further research into DM-ILD.
Collapse
Affiliation(s)
- Changjian Liu
- Department of Rheumatology, the Key Laboratory of Myositis, China-Japan Friendship Hospital, Beijing, People's Republic of China
| | - Yongpeng Ge
- Department of Rheumatology, the Key Laboratory of Myositis, China-Japan Friendship Hospital, Beijing, People's Republic of China
| |
Collapse
|
5
|
Yang W, Jin M, Gu Y, Zhao X, Zhu L, He S, Wang H, Ding X, Wang B, Jiang T, Xiao Y, Zhou G, Huang J, Zhang Y. Intracellular osteopontin potentiates the immunosuppressive activity of mesenchymal stromal cells. Stem Cell Res Ther 2024; 15:366. [PMID: 39407354 PMCID: PMC11475537 DOI: 10.1186/s13287-024-03979-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Accepted: 10/05/2024] [Indexed: 10/19/2024] Open
Abstract
INTRODUCTION Mesenchymal stromal cell (MSC)-based cell therapy is a promising approach for various inflammatory disorders based on their immunosuppressive capacity. Osteopontin (OPN) regulates several cellular functions including tissue repair, bone metabolism and immune reaction. However, the biological function of OPN in regulating the immunosuppressive capacity of MSCs remains elusive. OBJECTIVES This study aims to highlight the underlying mechanism of the proinflammatory cytokines affect the therapeutic ability of MSCs through OPN. METHODS MSCs in response to the proinflammatory cytokines were collected to determine the expression profile of OPN. In vitro T-cell proliferation assays and gene editing were performed to check the role and mechanisms of OPN in regulating the immunosuppressive capacity of MSCs. Inflammatory disease mouse models were established to evaluate the effect of OPN on improving MSC-based immunotherapy. RESULTS We observed that OPN, including its two isoforms iOPN and sOPN, was downregulated in MSCs upon proinflammatory cytokine stimulation. Interestingly, iOPN, but not sOPN, greatly enhanced the immunosuppressive activity of MSCs on T-cell proliferation and thus alleviated the inflammatory pathologies of hepatitis and colitis. Mechanistically, iOPN interacted with STAT1 and mediated its deubiquitination, thereby inducing the master immunosuppressive mediator inducible nitric oxide synthase (iNOS) in MSCs. In addition, iOPN expression was directly downregulated by activated STAT1, which formed a negative feedback loop to restrain MSC immunosuppressive capacity. CONCLUSION Our findings demonstrated that iOPN expression modulation in MSCs is a novel strategy to improve MSC-based immunotherapy.
Collapse
Affiliation(s)
- Wanlin Yang
- Department of Gastrointestinal Surgery, The Affiliated Changshu Hospital of Nantong University, Changshu, China
- Shanghai Lung Cancer Center, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Min Jin
- Department of Gastrointestinal Surgery, The Affiliated Changshu Hospital of Nantong University, Changshu, China
- Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yuting Gu
- Department of Gastrointestinal Surgery, The Affiliated Changshu Hospital of Nantong University, Changshu, China
- Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiaonan Zhao
- Children's Hospital of Soochow University, Institutes for Translational Medicine, State Key Laboratory of Radiation Medicine and Protection, Medical College of Soochow University, Soochow University, Suzhou, China
| | - Lingqiao Zhu
- Children's Hospital of Soochow University, Institutes for Translational Medicine, State Key Laboratory of Radiation Medicine and Protection, Medical College of Soochow University, Soochow University, Suzhou, China
| | - Shan He
- Children's Hospital of Soochow University, Institutes for Translational Medicine, State Key Laboratory of Radiation Medicine and Protection, Medical College of Soochow University, Soochow University, Suzhou, China
| | - Hui Wang
- Shanghai Lung Cancer Center, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xinyuan Ding
- Gusu College, Nanjing Medical University, Nanjing, China
| | - Bei Wang
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, Shanghai, China
| | - Tingwang Jiang
- Department of Gastrointestinal Surgery, The Affiliated Changshu Hospital of Nantong University, Changshu, China
| | - Yichuan Xiao
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, Shanghai, China
| | - Guoqiang Zhou
- Department of Gastrointestinal Surgery, The Affiliated Changshu Hospital of Nantong University, Changshu, China.
- Gusu College, Nanjing Medical University, Nanjing, China.
| | - Jiefang Huang
- Department of Gastrointestinal Surgery, The Affiliated Changshu Hospital of Nantong University, Changshu, China.
- Gusu College, Nanjing Medical University, Nanjing, China.
| | - Yanyun Zhang
- Department of Gastrointestinal Surgery, The Affiliated Changshu Hospital of Nantong University, Changshu, China.
- Children's Hospital of Soochow University, Institutes for Translational Medicine, State Key Laboratory of Radiation Medicine and Protection, Medical College of Soochow University, Soochow University, Suzhou, China.
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, Shanghai, China.
| |
Collapse
|
6
|
Levati L, Tabolacci C, Facchiano A, Facchiano F, Alvino E, Antonini Cappellini GC, Scala E, Bonmassar L, Caporali S, Lacal PM, Bresin A, De Galitiis F, Russo G, D'Atri S. Circulating interleukin-8 and osteopontin are promising biomarkers of clinical outcomes in advanced melanoma patients treated with targeted therapy. J Exp Clin Cancer Res 2024; 43:226. [PMID: 39143551 PMCID: PMC11325673 DOI: 10.1186/s13046-024-03151-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Accepted: 08/04/2024] [Indexed: 08/16/2024] Open
Abstract
BACKGROUND Circulating cytokines can represent non-invasive biomarkers to improve prediction of clinical outcomes of cancer patients. Here, plasma levels of IL-8, CCL4, osteopontin, LIF and BDNF were determined at baseline (T0), after 2 months of therapy (T2) and, when feasible, at progression (TP), in 70 melanoma patients treated with BRAF and MEK inhibitors. The association of baseline cytokine levels with clinical response, progression-free survival (PFS) and overall survival (OS) was evaluated. METHODS Cytokine concentrations were measured using the xMAP technology. Their ability to discriminate between responding (Rs) and non-responding (NRs) patients was assessed by Receiver Operating Characteristics analysis. PFS and OS were estimated with the Kaplan-Meier method. The Cox proportional hazard model was used in the univariate and multivariate analyses to estimate crude and adjusted hazard ratios with 95% confidence intervals. RESULTS CCL4 and LIF were undetectable in the majority of samples. The median osteopontin concentration at T0 and T2 was significantly higher in NRs than in Rs. The median T0 and T2 values of IL-8 were also higher in NRs than in Rs, although the statistical significance was not reached. No differences were detected for BDNF. In 39 Rs with matched T0, T2, and TP samples, osteopontin and IL-8 significantly decreased from T0 to T2 and rose again at TP, while BDNF levels remained unchanged. In NRs, none of the cytokines showed a significant decrease at T2. Only osteopontin demonstrated a good ability to discriminate between Rs and NRs. A high IL-8 T0 level was associated with significantly shorter PFS and OS and higher risk of progression and mortality, and remained an independent negative prognostic factor for OS in multivariate analysis. An elevated osteopontin T0 concentration was also significantly associated with worse OS and increased risk of death. Patients with high IL-8 and high osteopontin showed the lowest PFS and OS, and in multivariate analysis this cytokine combination remained independently associated with a three- to six-fold increased risk of mortality. CONCLUSION Circulating IL-8 and osteopontin appear useful biomarkers to refine prognosis evaluation of patients undergoing targeted therapy, and deserve attention as potential targets to improve its clinical efficacy.
Collapse
Affiliation(s)
- Lauretta Levati
- Laboratory of Molecular Oncology, Istituto Dermopatico Dell'Immacolata, IDI-IRCCS, Via Dei Monti Di Creta 104, 00167, Rome, Italy
| | - Claudio Tabolacci
- Department of Oncology and Molecular Medicine, Istituto Superiore Di Sanità, Viale Regina Elena 299, 00161, Rome, Italy
- Present Address: Research Coordination and Support Service, Istituto Superiore Di Sanità, Viale Regina Elena 299, 00161, Rome, Italy
| | - Antonio Facchiano
- Laboratory of Molecular Oncology, Istituto Dermopatico Dell'Immacolata, IDI-IRCCS, Via Dei Monti Di Creta 104, 00167, Rome, Italy
| | - Francesco Facchiano
- Department of Oncology and Molecular Medicine, Istituto Superiore Di Sanità, Viale Regina Elena 299, 00161, Rome, Italy
| | - Ester Alvino
- Institute of Translational Pharmacology, National Council of Research, Via Fosso del Cavaliere 100, 00133, Rome, Italy
| | - Gian Carlo Antonini Cappellini
- Department of Oncology and Dermatological Oncology, Istituto Dermopatico Dell'Immacolata, IDI-IRCCS, Via Dei Monti Di Creta 104, 00167, Rome, Italy
- Present Address: UOC Oncologia, Interpresidio ASL RM2, Via Dei Monti Tiburtini 387, 00157, Rome, Italy
| | - Enrico Scala
- Clinical and Laboratory Molecular Allergy Unit, Istituto Dermopatico Dell'Immacolata, IDI-IRCCS, Via Dei Monti Di Creta 104, 00167, Rome, Italy
| | - Laura Bonmassar
- Laboratory of Molecular Oncology, Istituto Dermopatico Dell'Immacolata, IDI-IRCCS, Via Dei Monti Di Creta 104, 00167, Rome, Italy
| | - Simona Caporali
- Laboratory of Molecular Oncology, Istituto Dermopatico Dell'Immacolata, IDI-IRCCS, Via Dei Monti Di Creta 104, 00167, Rome, Italy
- Present Address: Regional Transplant Center Lazio (CRTL), San Camillo Hospital, Circonvallazione Gianicolense 87, 00152, Rome, Italy
| | - Pedro Miguel Lacal
- Laboratory of Molecular Oncology, Istituto Dermopatico Dell'Immacolata, IDI-IRCCS, Via Dei Monti Di Creta 104, 00167, Rome, Italy
| | - Antonella Bresin
- Laboratory of Molecular Oncology, Istituto Dermopatico Dell'Immacolata, IDI-IRCCS, Via Dei Monti Di Creta 104, 00167, Rome, Italy
| | - Federica De Galitiis
- Department of Oncology and Dermatological Oncology, Istituto Dermopatico Dell'Immacolata, IDI-IRCCS, Via Dei Monti Di Creta 104, 00167, Rome, Italy
| | - Giandomenico Russo
- Laboratory of Molecular Oncology, Istituto Dermopatico Dell'Immacolata, IDI-IRCCS, Via Dei Monti Di Creta 104, 00167, Rome, Italy
| | - Stefania D'Atri
- Laboratory of Molecular Oncology, Istituto Dermopatico Dell'Immacolata, IDI-IRCCS, Via Dei Monti Di Creta 104, 00167, Rome, Italy.
| |
Collapse
|
7
|
Śmieszek A, Marcinkowska K, Małas Z, Sikora M, Kępska M, Nowakowska BA, Deperas M, Smyk M, Rodriguez-Galindo C, Raciborska A. Identification and characterization of stromal-like cells with CD207 +/low CD1a +/low phenotype derived from histiocytic lesions - a perspective in vitro model for drug testing. BMC Cancer 2024; 24:105. [PMID: 38342891 PMCID: PMC10860276 DOI: 10.1186/s12885-023-11807-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Accepted: 12/28/2023] [Indexed: 02/13/2024] Open
Abstract
BACKGROUND Histiocytoses are rare disorders manifested by increased proliferation of pathogenic myeloid cells sharing histological features with macrophages or dendritic cells and accumulating in various organs, i.a., bone and skin. Pre-clinical in vitro models that could be used to determine molecular pathways of the disease are limited, hence research on histiocytoses is challenging. The current study compares cytophysiological features of progenitor, stromal-like cells derived from histiocytic lesions (sl-pHCs) of three pediatric patients with different histiocytoses types and outcomes. The characterized cells may find potential applications in drug testing. METHODS Molecular phenotype of the cells, i.e. expression of CD1a and CD207 (langerin), was determined using flow cytometry. Cytogenetic analysis included GTG-banded metaphases and microarray (aCGH) evaluation. Furthermore, the morphology and ultrastructure of cells were evaluated using a confocal and scanning electron microscope. The microphotographs from the confocal imaging were used to reconstruct the mitochondrial network and its morphology. Basic cytophysiological parameters, such as viability, mitochondrial activity, and proliferation, were analyzed using multiple cellular assays, including Annexin V/7-AAD staining, mitopotential analysis, BrdU test, clonogenicity analysis, and distribution of cells within the cell cycle. Biomarkers potentially associated with histiocytoses progression were determined using RT-qPCR at mRNA, miRNA and lncRNA levels. Intracellular accumulation of histiocytosis-specific proteins was detected with Western blot. Cytotoxicyty and IC50 of vemurafenib and trametinib were determined with MTS assay. RESULTS Obtained cellular models, i.e. RAB-1, HAN-1, and CHR-1, are heterogenic in terms of molecular phenotype and morphology. The cells express CD1a/CD207 markers characteristic for dendritic cells, but also show intracellular accumulation of markers characteristic for cells of mesenchymal origin, i.e. vimentin (VIM) and osteopontin (OPN). In subsequent cultures, cells remain viable and metabolically active, and the mitochondrial network is well developed, with some distinctive morphotypes noted in each cell line. Cell-specific transcriptome profile was noted, providing information on potential new biomarkers (non-coding RNAs) with diagnostic and prognostic features. The cells showed different sensitivity to vemurafenib and trametinib. CONCLUSION Obtained and characterized cellular models of stromal-like cells derived from histiocytic lesions can be used for studies on histiocytosis biology and drug testing.
Collapse
Affiliation(s)
- Agnieszka Śmieszek
- Department of Pharmacology and Toxicology, Faculty of Veterinary Medicine, Wroclaw University of Environmental and Life Sciences, Norwida 31, 50-375, Wroclaw, Poland.
| | - Klaudia Marcinkowska
- Department of Experimental Biology, Faculty of Biology and Animal Science, Wroclaw University of Environmental and Life Sciences, Norwida 27B, 50-375, Wroclaw, Poland
| | - Zofia Małas
- Department of Oncology and Surgical Oncology for Children and Youth, Institute of Mother and Child, Kasprzaka 17a, 01-211, Warsaw, Poland
| | - Mateusz Sikora
- Department of Experimental Biology, Faculty of Biology and Animal Science, Wroclaw University of Environmental and Life Sciences, Norwida 27B, 50-375, Wroclaw, Poland
| | - Martyna Kępska
- Department of Experimental Biology, Faculty of Biology and Animal Science, Wroclaw University of Environmental and Life Sciences, Norwida 27B, 50-375, Wroclaw, Poland
| | - Beata A Nowakowska
- Medical Genetics Department, Cytogenetics Laboratory, Institute of Mother and Child, Kasprzaka 17a, 01-211, Warsaw, Poland
| | - Marta Deperas
- Medical Genetics Department, Cytogenetics Laboratory, Institute of Mother and Child, Kasprzaka 17a, 01-211, Warsaw, Poland
| | - Marta Smyk
- Medical Genetics Department, Cytogenetics Laboratory, Institute of Mother and Child, Kasprzaka 17a, 01-211, Warsaw, Poland
| | | | - Anna Raciborska
- Department of Oncology and Surgical Oncology for Children and Youth, Institute of Mother and Child, Kasprzaka 17a, 01-211, Warsaw, Poland.
| |
Collapse
|
8
|
Cohen R, Mahlab-Guri K, Atali M, Elbirt D. Viruses and celiac disease: what do we know ? Clin Exp Med 2023; 23:2931-2939. [PMID: 37103650 PMCID: PMC10134706 DOI: 10.1007/s10238-023-01070-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Accepted: 04/05/2023] [Indexed: 04/28/2023]
Abstract
The aim of this review is to provide a comprehensive overview about the link between viruses and celiac disease. A systematic search on PubMed, Embase, and Scopus was conducted on March 07, 2023. The reviewers independently selected the articles and chose which articles to include. The review is a textual systemic review, and all relevant articles were included based on title and abstract. If there was a disagreement between the reviewers, they came to a consensus during deliberation sessions. A total of 178 articles were selected for the review and read in full; only part of them was retained. We found studies between celiac disease and 12 different viruses. Some of the studies were done only on small groups. Most studies were on pediatric population. Evidence for an association was found with several viruses (trigger or protective). It seems that only a part of the viruses could induce the disease. Several points are important to keep in mind: firstly, simple mimicry or that the virus induces a high level of TGA is not sufficient to promote the disease. Secondly, inflammatory background is necessary to induce CD with virus. Thirdly, IFN type 1 seems to have an important role. Some of the viruses are potential or known triggers like enteroviruses, rotaviruses, reoviruses, and influenza. Further studies are needed to better understand the role of viruses in celiac disease to better treat and prevent the disease.
Collapse
Affiliation(s)
- Ramon Cohen
- Internal Department B, Kaplan Medical Center, Rehovot, Israel.
- Department of Clinical Immunology Allergy and AIDS, Kaplan Medical Center, Rehovot, Israel.
| | - Keren Mahlab-Guri
- Department of Clinical Immunology Allergy and AIDS, Kaplan Medical Center, Rehovot, Israel
| | - Malka Atali
- Internal Department B, Kaplan Medical Center, Rehovot, Israel
| | - Daniel Elbirt
- Department of Clinical Immunology Allergy and AIDS, Kaplan Medical Center, Rehovot, Israel
| |
Collapse
|
9
|
Sumioka T, Matsumoto KI, Reinach PS, Saika S. Tenascins and osteopontin in biological response in cornea. Ocul Surf 2023; 29:131-149. [PMID: 37209968 DOI: 10.1016/j.jtos.2023.05.005] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Revised: 05/01/2023] [Accepted: 05/16/2023] [Indexed: 05/22/2023]
Abstract
The structural composition, integrity and regular curvature of the cornea contribute to the maintenance of its transparency and vision. Disruption of its integrity caused by injury results in scarring, inflammation and neovascularization followed by losses in transparency. These sight compromising effects is caused by dysfunctional corneal resident cell responses induced by the wound healing process. Upregulation of growth factors/cytokines and neuropeptides affect development of aberrant behavior. These factors trigger keratocytes to first transform into activated fibroblasts and then to myofibroblasts. Myofibroblasts express extracellular matrix components for tissue repair and contract the tissue to facilitate wound closure. Proper remodeling following primary repair is critical for restoration of transparency and visual function. Extracellular matrix components contributing to the healing process are divided into two groups; a group of classical tissue structural components and matrix macromolecules that modulate cell behaviors/activities besides being integrated into the matrix structure. The latter components are designated as matricellular proteins. Their functionality is elicited through mechanisms which modulate the scaffold integrity, cell behaviors, activation/inactivation of either growth factors or cytoplasmic signaling regulation. We discuss here the functional roles of matricellular proteins in mediating injury-induced corneal tissue repair. The roles are described of major matricellular proteins, which include tenascin C, tenascin X and osteopontin. Focus is directed towards dealing with their roles in modulating individual activities of wound healing-related growth factors, e. g., transforming growth factor β (TGF β). Modulation of matricellular protein functions could encompass a potential novel strategy to improve the outcome of injury-induced corneal wound healing.
Collapse
Affiliation(s)
- Takayoshi Sumioka
- Department of Ophthalmology, Wakayama Medical University School of Medicine, 811-1 Kimiidera, 641-0012, Japan.
| | - Ken-Ichi Matsumoto
- Department of Biosignaling and Radioisotope Experiment, Interdisciplinary Center for Science Research, Head Office for Research and Academic Information, Shimane University, 89-1 Enya-cho, Izumo, 693-8501, Japan
| | - Peter Sol Reinach
- Department of Biological. Sciences SUNY Optometry, New York, NY, 10036, USA
| | - Shizuya Saika
- Department of Ophthalmology, Wakayama Medical University School of Medicine, 811-1 Kimiidera, 641-0012, Japan
| |
Collapse
|
10
|
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: 6.0] [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
|
11
|
Liu Y, Fu L, Liu Z. The Role and Clinical Relevance of Osteopontin in Allergic Airway Diseases. J Clin Med 2023; 12:jcm12062433. [PMID: 36983433 PMCID: PMC10057512 DOI: 10.3390/jcm12062433] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Revised: 03/06/2023] [Accepted: 03/15/2023] [Indexed: 03/30/2023] Open
Abstract
The airway epithelium is exposed to numerous external irritants including infectious agents, environmental allergens, and atmospheric pollutants, releasing epithelial cytokines including thymic stromal lymphopoietin (TSLP), IL-33, and IL-25 and initiating downstream type 2 (IL-4, IL-13, and IL-5) and IgE-driven pathways. These pathways trigger the initiation and progression of allergic airway diseases, including chronic rhinosinusitis with nasal polyps (CRSwNP), allergic rhinitis (AR), and allergic asthma. However, the use of biological agents that target downstream cytokines, such as IL-5, IL-4, and IL-13 receptors and IgE, might not be sufficient to manage some patients successfully. Instead of blocking downstream cytokines, targeting upstream epithelial cytokines has been proposed to address the complex immunologic networks associated with allergic airway diseases. Osteopontin (OPN), an extracellular matrix glyco-phosphoprotein, is a key mediator involved in Th1-related diseases, including systemic lupus erythematosus, multiple sclerosis, inflammatory bowel disease, and rheumatoid arthritis. Emerging evidence, including ours, indicates that epithelial-cell-derived OPN also plays an essential role in Th2-skewed airway diseases, including CRSwNP, AR, and allergic asthma involving the Th17 response. Therefore, we reviewed the current knowledge of epithelial-cell-derived OPN in the pathogenesis of three type-2-biased airway diseases and provided a direction for its future investigation and clinical relevance.
Collapse
Affiliation(s)
- Yang Liu
- Department of Otolaryngology-Head and Neck Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
- Institute of Allergy and Clinical Immunology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
- Hubei Clinical Research Center for Nasal Inflammatory Diseases, Wuhan 430030, China
| | - Li Fu
- Department of Otolaryngology-Head and Neck Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
- Institute of Allergy and Clinical Immunology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
- Hubei Clinical Research Center for Nasal Inflammatory Diseases, Wuhan 430030, China
| | - Zheng Liu
- Department of Otolaryngology-Head and Neck Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
- Institute of Allergy and Clinical Immunology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
- Hubei Clinical Research Center for Nasal Inflammatory Diseases, Wuhan 430030, China
| |
Collapse
|
12
|
Friedlová N, Zavadil Kokáš F, Hupp TR, Vojtěšek B, Nekulová M. IFITM protein regulation and functions: Far beyond the fight against viruses. Front Immunol 2022; 13:1042368. [PMID: 36466909 PMCID: PMC9716219 DOI: 10.3389/fimmu.2022.1042368] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Accepted: 10/27/2022] [Indexed: 07/30/2023] Open
Abstract
Interferons (IFNs) are important cytokines that regulate immune responses through the activation of hundreds of genes, including interferon-induced transmembrane proteins (IFITMs). This evolutionarily conserved protein family includes five functionally active homologs in humans. Despite the high sequence homology, IFITMs vary in expression, subcellular localization and function. The initially described adhesive and antiproliferative or pro-oncogenic functions of IFITM proteins were diluted by the discovery of their antiviral properties. The large set of viruses that is inhibited by these proteins is constantly expanding, as are the possible mechanisms of action. In addition to their beneficial antiviral effects, IFITM proteins are often upregulated in a broad spectrum of cancers. IFITM proteins have been linked to most hallmarks of cancer, including tumor cell proliferation, therapeutic resistance, angiogenesis, invasion, and metastasis. Recent studies have described the involvement of IFITM proteins in antitumor immunity. This review summarizes various levels of IFITM protein regulation and the physiological and pathological functions of these proteins, with an emphasis on tumorigenesis and antitumor immunity.
Collapse
Affiliation(s)
- Nela Friedlová
- Research Centre for Applied Molecular Oncology, Masaryk Memorial Cancer Institute, Brno, Czechia
- Department of Experimental Biology, Faculty of Science, Masaryk University, Brno, Czechia
| | - Filip Zavadil Kokáš
- Research Centre for Applied Molecular Oncology, Masaryk Memorial Cancer Institute, Brno, Czechia
| | - Ted R. Hupp
- Research Centre for Applied Molecular Oncology, Masaryk Memorial Cancer Institute, Brno, Czechia
- Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, United Kingdom
| | - Bořivoj Vojtěšek
- Research Centre for Applied Molecular Oncology, Masaryk Memorial Cancer Institute, Brno, Czechia
| | - Marta Nekulová
- Research Centre for Applied Molecular Oncology, Masaryk Memorial Cancer Institute, Brno, Czechia
| |
Collapse
|
13
|
Jia Q, Huang Z, Wang G, Sun X, Wu Y, Yang B, Yang T, Liu J, Li P, Li J. Osteopontin: An important protein in the formation of kidney stones. Front Pharmacol 2022; 13:1036423. [PMID: 36452224 PMCID: PMC9703462 DOI: 10.3389/fphar.2022.1036423] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2022] [Accepted: 10/26/2022] [Indexed: 09/10/2024] Open
Abstract
The incidence of kidney stones averages 10%, and the recurrence rate of kidney stones is approximately 10% at 1 year, 35% at 5 years, 50% at 10 years, and 75% at 20 years. However, there is currently a lack of good medicines for the prevention and treatment of kidney stones. Osteopontin (OPN) is an important protein in kidney stone formation, but its role is controversial, with some studies suggesting that it inhibits stone formation, while other studies suggest that it can promote stone formation. OPN is a highly phosphorylated protein, and with the deepening of research, there is growing evidence that it promotes stone formation, and the phosphorylated protein is believed to have adhesion effect, promote stone aggregation and nucleation. In addition, OPN is closely related to immune cell infiltration, such as OPN as a pro-inflammatory factor, which can activate mast cells (degranulate to release various inflammatory factors), macrophages (differentiated into M1 macrophages), and T cells (differentiated into T1 cells) etc., and these inflammatory cells play a role in kidney damage and stone formation. In short, OPN mainly exists in the phosphorylated form in kidney stones, plays an important role in the formation of stones, and may be an important target for drug therapy of kidney stones.
Collapse
Affiliation(s)
- Qingxia Jia
- The Department of Urology, The Second Affiliated Hospital of Kunming Medical University, Kunming, China
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, China
| | - Ziye Huang
- The Department of Urology, The Second Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Guang Wang
- The Department of Urology, The Second Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Xia Sun
- The Department of Urology, The Second Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Yuyun Wu
- The Department of Urology, The Second Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Bowei Yang
- The Department of Urology, The Second Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Tongxin Yang
- The Department of Urology, The Second Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Jianhe Liu
- The Department of Urology, The Second Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Pei Li
- The Department of Urology, The Second Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Jiongming Li
- The Department of Urology, The Second Affiliated Hospital of Kunming Medical University, Kunming, China
| |
Collapse
|
14
|
Jiang ZF, Wu W, Hu HB, Li ZY, Zhong M, Zhang L. P2X7 receptor as the regulator of T-cell function in intestinal barrier disruption. World J Gastroenterol 2022; 28:5265-5279. [PMID: 36185635 PMCID: PMC9521516 DOI: 10.3748/wjg.v28.i36.5265] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/12/2022] [Revised: 06/20/2022] [Accepted: 09/01/2022] [Indexed: 02/06/2023] Open
Abstract
The intestinal mucosa is a highly compartmentalized structure that forms a direct barrier between the host intestine and the environment, and its dysfunction could result in a serious disease. As T cells, which are important components of the mucosal immune system, interact with gut microbiota and maintain intestinal homeostasis, they may be involved in the process of intestinal barrier dysfunction. P2X7 receptor (P2X7R), a member of the P2X receptors family, mediates the effects of extracellular adenosine triphosphate and is expressed by most innate or adaptive immune cells, including T cells. Current evidence has demonstrated that P2X7R is involved in inflammation and mediates the survival and differentiation of T lymphocytes, indicating its potential role in the regulation of T cell function. In this review, we summarize the available research about the regulatory role and mechanism of P2X7R on the intestinal mucosa-derived T cells in the setting of intestinal barrier dysfunction.
Collapse
Affiliation(s)
- Zhi-Feng Jiang
- Center of Emergency & Intensive Care Unit, Jinshan Hospital of Fudan University, Shanghai 201508, China
| | - Wei Wu
- Department of Critical Care Medicine, Zhongshan Hospital of Fudan University, Shanghai 200032, China
| | - Han-Bing Hu
- Center of Emergency & Intensive Care Unit, Jinshan Hospital of Fudan University, Shanghai 201508, China
| | - Zheng-Yang Li
- Department of Gastroenterology, Jinshan Hospital of Fudan University, Shanghai 201508, China
| | - Ming Zhong
- Department of Critical Care Medicine, Zhongshan Hospital of Fudan University, Shanghai 200032, China
| | - Lin Zhang
- Center of Emergency & Intensive Care Unit, Jinshan Hospital of Fudan University, Shanghai 201508, China
| |
Collapse
|
15
|
A Four-Gene Signature Associated with Radioresistance in Head and Neck Squamous Cell Carcinoma Identified by Text Mining and Data Analysis. COMPUTATIONAL AND MATHEMATICAL METHODS IN MEDICINE 2022; 2022:5693806. [PMID: 36203528 PMCID: PMC9532131 DOI: 10.1155/2022/5693806] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Accepted: 09/12/2022] [Indexed: 12/24/2022]
Abstract
Purpose Head and neck squamous cell carcinoma (HNSCC) is the sixth leading cancer globally, and radiotherapy plays a crucial part in its treatment. This study was designed to identify potential genes related to radiation resistance in HNSCC. Method We first used text mining to obtain common genes related to radiotherapy resistance and HNSCC in published articles. Functional enrichment analyses were conducted to identify the significantly enriched pathways and genes. Protein and protein interactions were performed, and the most significant gene modules were determined; then, genes in the gene modules were validated at transcriptional levels and overall survival. Gene set variation analysis (GSVA) score was calculated, and the association between GSVA score and survival/pathway was estimated. Immune cell infiltration, methylation, and genetic alteration analysis of these genes was conducted in HNSCC patients. Finally, potential sensitive anticancer drugs related to target genes were obtained. Result We identified 583 common genes through text mining. After further validation, a four-gene signature (EPHB2, SPP1, SERPINE1, and VEGFC) was constructed. The patients with higher GSVA scores have a worse prognosis than those with lower GSVA scores. Differences in methylation of these four genes in HNSCC tumor tissue and normal tissue were compared, with higher methylation levels of EBPH2 and SPP1 in normal tissue and higher methylation levels of SERPINE1 in the tumor. Immune cell infiltration revealed that the increased expression of these genes was closely related to the infiltration level of CD4+ T cell, neutrophil, macrophage, and dendritic cell. Thirty drugs, including 22 positively and eight negatively correlated drugs that most correlated with related genes, were available for treating HNSCC. Conclusion In this study, we identified four potential genes as well as corresponding drugs that might be related to radioresistance in HNSCC patients. These candidate genes may provide a promising avenue to further elevate radiotherapy efficacy.
Collapse
|
16
|
Jámbor K, Koroknai V, Kiss T, Szász I, Pikó P, Balázs M. Gene Expression Patterns of Osteopontin Isoforms and Integrins in Malignant Melanoma. PATHOLOGY AND ONCOLOGY RESEARCH 2022; 28:1610608. [PMID: 36091936 PMCID: PMC9448871 DOI: 10.3389/pore.2022.1610608] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Accepted: 07/11/2022] [Indexed: 11/30/2022]
Abstract
Osteopontin (OPN) is a multifunctional glycoprotein that physiologically interacts with different types of integrins. It is considered to be a possible prognostic biomarker in certain tumor types; however, various splicing isoforms exist, which have not been investigated in melanoma. We aimed to define the relative expression pattern of five OPN isoforms and clarify the prognostic significance of the splice variants in melanoma. We also aimed to investigate the expression pattern of eight integrins in the same tumors. Gene expression analyses revealed that the relative expression of OPNa, OPNb, and OPNc is significantly higher in metastatic tumors compared to primary lesions (p < 0.01), whereas the expression of OPN4 and OPN5 was low in both. The more aggressive nodular melanomas had higher expression levels compared to the superficial spreading subtype (p ≤ 0.05). The relative expression of the eight tested integrins was low, with only the expression of ITGB3 being detectable in nodular melanoma (Medianlog2 = 1.274). A positive correlation was found between Breslow thickness and the expression of OPNc variant, whereby thicker tumors (>4 mm) had significantly higher expression (p ≤ 0.05). The Breslow thickness was negatively correlated with the expression of OPN4, and similarly with ITGA2. OPNc also exhibited significant positive correlation with the presence of metastasis. Our data show that high expression of OPNa, OPNb, and especially OPNc and low expression of OPN4 and ITGA2 are associated with an advanced stage of tumor progression and poor prognosis in melanoma.
Collapse
Affiliation(s)
- Krisztina Jámbor
- Doctoral School of Health Sciences, University of Debrecen, Debrecen, Hungary
- Department of Public Health and Epidemiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Viktória Koroknai
- Department of Public Health and Epidemiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
- ELKH-DE Public Health Research Group, University of Debrecen, Debrecen, Hungary
| | - Tímea Kiss
- Department of Public Health and Epidemiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - István Szász
- Department of Public Health and Epidemiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
- ELKH-DE Public Health Research Group, University of Debrecen, Debrecen, Hungary
| | - Péter Pikó
- Department of Public Health and Epidemiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
- ELKH-DE Public Health Research Group, University of Debrecen, Debrecen, Hungary
| | - Margit Balázs
- Department of Public Health and Epidemiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
- ELKH-DE Public Health Research Group, University of Debrecen, Debrecen, Hungary
- *Correspondence: Margit Balázs,
| |
Collapse
|
17
|
Hernández-Bazán S, Mata-Espinosa D, Lozano-Ordaz V, Ramos-Espinosa O, Barrios-Payán J, López-Casillas F, Hernández-Pando R. Immune regulatory effect of osteopontin gene therapy in a murine model of multi-drug resistant pulmonary tuberculosis. Hum Gene Ther 2022; 33:1037-1051. [PMID: 35615876 DOI: 10.1089/hum.2022.030] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Abstract
Tuberculosis (TB) has been for many years a major public health problem since treatment is long and sometimes ineffective favoring the increase of multi-drug-resistant mycobacteria (MDR). Gene therapy is a novel and effective tool to regulate immune responses. In this study we evaluated the therapeutic effect of an adenoviral vector codifying osteopontin (AdOPN), a molecule known for their roles to favour Th1 and Th17 type-cytokine expression which are crucial in TB containment. A single-dose of AdOPN administration in BALB/c mice suffering late progressive pulmonary MDR-TB, produced significant lower bacterial load and pneumonia, due to higher expression of IFN-γ, IL-12 and IL-17 in coexistence with increase of granulomas in number and size, resulting in higher survival, in contrast with mice treated with the control adenovirus that codify the green fluorescent protein (AdGFP). Combined therapy of AdOPN with a regimen of 2nd line antibiotics produced a better control of bacterial load in lung during the first days of treatment, suggesting that AdOPN can shorten chemotherapy. Taken together, gene therapy with AdOPN leads to higher immune responses against TB infection, resulting in a new potential treatment against pulmonary TB that can co-adjuvant chemotherapy.
Collapse
Affiliation(s)
- Sujhey Hernández-Bazán
- Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubiran, 42559, Department of Pathology, Experimental Pathology Section, Tlalpan, CDMX, Mexico;
| | - Dulce Mata-Espinosa
- Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubiran, 42559, Department of Pathology, Experimental Pathology Section, Tlalpan, CDMX, Mexico;
| | - Vasti Lozano-Ordaz
- Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubiran, 42559, Department of Pathology, Experimental Pathology Section, Tlalpan, CDMX, Mexico;
| | - Octavio Ramos-Espinosa
- Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubiran, 42559, Department of Pathology, Experimental Pathology Section, Tlalpan, CDMX, Mexico;
| | - Jorge Barrios-Payán
- Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubiran, 42559, Department of Pathology, Experimental Pathology Section, Tlalpan, CDMX, Mexico;
| | - Fernando López-Casillas
- Universidad Nacional Autónoma de México Instituto de Fisiología Celular, 61739, Department of Cellular and Developmental Biology, Coyoacán, CDMX, Mexico;
| | - Rogelio Hernández-Pando
- Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubiran, 42559, Department of Pathology, Experimental Pathology Section, Tlalpan, CDMX, Mexico;
| |
Collapse
|
18
|
Wang M, Sun X, Xin H, Wen Z, Cheng Y. SPP1 promotes radiation resistance through JAK2/STAT3 pathway in esophageal carcinoma. Cancer Med 2022; 11:4526-4543. [PMID: 35593388 PMCID: PMC9741975 DOI: 10.1002/cam4.4840] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Revised: 04/28/2022] [Accepted: 05/02/2022] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Therapeutic resistance to radiotherapy is one of the major obstacles in clinical practice that significantly affect the therapeutic efficiency and prognosis of human esophageal carcinoma (ESCA). Thus, it is critical to understand the molecular mechanisms of radiation resistance in ESCA. Secreted phosphoprotein 1 (SPP1) plays an essential role in various human cancers, but its role in radiation resistance remains unclear. METHOD Cell culture and transfection; Cell Counting Kit-8 (CCK-8) assays; EdU incorporation assays; Patient sample collection and medical records review; Transwell assays; Colony formation assays; Wound healing assays; Western blot; Immunofluorescence; Immunohistochemistry; Irradiation; Flow cytometry; Animal studies; Human Apoptosis Array Kit; Bioinformatics. RESULT In the current study, we reported the novel phenomenon that radiation-treated human ESCA cells upregulated SPP1 expression, which in turn contributed to the ESCA resistance to radiotherapy. We also reported the tumor-promoting effect of SPP1 in ESCA systematically and comprehensively. Furthermore, subsequent studies by knocking down or overexpressing SPP1 in human ESCA cells showed that SPP1 could facilitate the repair of DNA damage and the survival of tumor cells post-radiation in ESCA, which might contribute to the development of radiation resistance during the radiotherapy process. More detailed investigations on the downstream molecular pathway suggested that radiation could increase the phosphorylation level of JAK2 and STAT3 by increasing SPP1 expression. Further in vivo validation using a mouse ESCA xenograft model showed that SPP1 overexpression significantly increased tumor volume while either SPP1 knockdown or pharmacological inhibition of the JAK2-STAT3 pathway reduced tumor volume in a synergistic manner with radiotherapy. CONCLUSION Collectively, these findings suggested that the SPP1/JAK2/STAT3 axis is a critical player in ESCA progression and radiation resistance, which is a potential therapeutic target for combined therapy with the standard radiotherapy regimen to improve curative effect and increase patients' survival with ESCA.
Collapse
Affiliation(s)
- Meijie Wang
- Department of Radiation Oncology, Qilu Hospital, Cheeloo College of MedicineQilu Hospital, Cheeloo College of MedicineShandong UniversityJinanChina,Laboratory of Basic Medical Sciences, Qilu Hospital, Cheeloo College of MedicineShandong UniversityJinanChina
| | - Xiaozheng Sun
- Department of Radiation Oncology, Qilu Hospital, Cheeloo College of MedicineQilu Hospital, Cheeloo College of MedicineShandong UniversityJinanChina,Laboratory of Basic Medical Sciences, Qilu Hospital, Cheeloo College of MedicineShandong UniversityJinanChina
| | - Huixian Xin
- Department of Radiation Oncology, Qilu Hospital, Cheeloo College of MedicineQilu Hospital, Cheeloo College of MedicineShandong UniversityJinanChina,Laboratory of Basic Medical Sciences, Qilu Hospital, Cheeloo College of MedicineShandong UniversityJinanChina
| | - Zhihua Wen
- Department of Radiation Oncology, Qilu Hospital, Cheeloo College of MedicineQilu Hospital, Cheeloo College of MedicineShandong UniversityJinanChina
| | - Yufeng Cheng
- Department of Radiation Oncology, Qilu Hospital, Cheeloo College of MedicineQilu Hospital, Cheeloo College of MedicineShandong UniversityJinanChina
| |
Collapse
|
19
|
Horna P, King RL, Jevremovic D, Fajgenbaum DC, Dispenzieri A. The lymph node transcriptome of unicentric and idiopathic multicentric Castleman disease. Haematologica 2022; 108:207-218. [PMID: 35484648 PMCID: PMC9827154 DOI: 10.3324/haematol.2021.280370] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Indexed: 02/05/2023] Open
Abstract
Castleman disease is a polyclonal lymphoproliferative disorder characterized by unicentric or multicentric lymphadenopathy with characteristic histomorphological features, in addition to variable inflammatory symptomatology. The molecular mechanisms and etiologies of unicentric Castleman disease (UCD) and idiopathic multicentric Castleman disease (iMCD) are poorly understood, and identification of targetable disease mediators remains an unmet clinical need. We performed whole exome sequencing on lymph node biopsies from patients with UCD and iMCD and compared the transcriptomic profiles to that of benign control lymph nodes. We identified significantly upregulated genes in UCD (n=443), iMCD (n=316) or both disease subtypes (n=51) and downregulated genes in UCD (n=321), iMCD (n=105) or both (n=10). The transcriptomes of UCD and iMCD showed enrichment and upregulation of elements of the complement cascade. By immunohistochemistry, C4d deposits indicative of complement activation were found to be present in UCD and iMCD, mostly within abnormally regressed germinal centers, but also in association with plasma cell clusters, endothelial cells and stroma cell proliferations. Other enriched gene sets included collagen organization, S1P3 pathway and VEGFR pathway in UCD; and humoral response, oxidative phosphorylation and proteosome in iMCD. Analysis of cytokine transcripts showed upregulation of CXCL13 but not IL6 in UCD and iMCD. Among angiogenic mediators, the VEGFR1 ligand placental growth factor (PGF) was upregulated in both disease subtypes. We hereby report for the first time the whole lymph node transcriptomes of UCD and iMCD, underscoring findings that could aid in the discovery of targetable disease mediators.
Collapse
Affiliation(s)
- Pedro Horna
- Division of Hematopathology, Mayo Clinic, Rochester, MN,P. Horna
| | | | | | - David C. Fajgenbaum
- Center for Cytokine Storm Treatment & Laboratory, University of Pennsylvania, Philadelphia, PA
| | | |
Collapse
|
20
|
Higher Expression of SPP1 Predicts Poorer Survival Outcomes in Head and Neck Cancer. J Immunol Res 2022; 2021:8569575. [PMID: 34977258 PMCID: PMC8718292 DOI: 10.1155/2021/8569575] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Revised: 11/09/2021] [Accepted: 11/16/2021] [Indexed: 12/03/2022] Open
Abstract
Secreted phosphoprotein 1 (SPP1) participated in various biological processes in many cancers, including immune response, tumor progression, and prognosis. However, SPP1 in head and neck squamous cell carcinoma (HNSCC) remains unknown. Clinical-genetic data of HNSCC were obtained from The Cancer Genome Atlas (TCGA) database. The differential expression of SPP1 in HNSCC tissues and adjacent normal tissues was quantified by bioinformatics methods and verified by western blot and other differential biological methods. We concluded that SPP1 is significantly upregulated in tumor tissues and can become a prognostic biomarker for HNSCC.
Collapse
|
21
|
Marastoni D, Magliozzi R, Bolzan A, Pisani AI, Rossi S, Crescenzo F, Montemezzi S, Pizzini FB, Calabrese M. CSF Levels of CXCL12 and Osteopontin as Early Markers of Primary Progressive Multiple Sclerosis. NEUROLOGY-NEUROIMMUNOLOGY & NEUROINFLAMMATION 2021; 8:8/6/e1083. [PMID: 34588298 PMCID: PMC8482414 DOI: 10.1212/nxi.0000000000001083] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Accepted: 07/21/2021] [Indexed: 11/20/2022]
Abstract
Background and Objectives To evaluate the extent of intrathecal inflammation in patients with primary progressive MS (PPMS) at the time of diagnosis and to define markers and a specific inflammatory profile capable of distinguishing progressive from relapsing-remitting multiple sclerosis (RRMS). Methods Levels of 34 pro- and anti-inflammatory cytokines and chemokines in the CSF were evaluated at the diagnosis in 16 patients with PPMS and 80 with RRMS. All patients underwent clinical evaluation, including Expanded Disability Status Scale assessment and a 3T brain MRI to detect white matter and cortical lesion number and volume and global and regional cortical thickness. Results Higher levels of CXCL12 (odds ratio [OR] = 3.97, 95% CI [1.34–11.7]) and the monocyte-related osteopontin (OR = 2.24, 95% CI [1.01–4.99]) were detected in patients with PPMS, whereas levels of interleukin-10 (IL10) (OR = 0.28, 95% CI [0.09–0.96]) were significantly increased in those with RRMS. High CXCL12 levels were detected in patients with increased gray matter lesion number and volume (p = 0.001, r = 0.832 and r = 0.821, respectively). Pathway analysis confirmed the chronic inflammatory processes occurring in PPMS. Conclusions At the time of diagnosis, a specific CSF protein profile can recognize the presence of early intrathecal inflammatory processes, possibly stratifying PPMS with respect to RRMS. Elevated CSF levels of CXCL12 and osteopontin suggested a key role of brain innate immunity and glia activity in MS. These molecules could represent useful candidate markers of MS progression, with implications for the pathogenesis and treatment of progressive MS. Classification of Evidence This study provides Class III evidence that CXCL12 and monocyte-related osteopontin may be correlated with PPMS, and IL-10 may be related to RRMS. It is may be correlated due to Bonferroni correction negating the statistical correlations found in the study.
Collapse
Affiliation(s)
- Damiano Marastoni
- From the Department of Neurosciences (D.M., R.M., A.B., A.I.P., F.C., M.C.), Biomedicine and Movement Sciences, University of Verona; Department of Brain Sciences (R.M.), Imperial College London, Hammersmith Hospital, UK; Department of Oncology and Molecular Medicine (S.R.), Higher Institute of Health Care, Rome; Neuroradiology & Radiology Units (S.M.), Integrated University Hospital of Verona; and Radiology (F.B.P.), Department of Diagnostic and Public Health, Integrated University Hospital of Verona, Italy
| | - Roberta Magliozzi
- From the Department of Neurosciences (D.M., R.M., A.B., A.I.P., F.C., M.C.), Biomedicine and Movement Sciences, University of Verona; Department of Brain Sciences (R.M.), Imperial College London, Hammersmith Hospital, UK; Department of Oncology and Molecular Medicine (S.R.), Higher Institute of Health Care, Rome; Neuroradiology & Radiology Units (S.M.), Integrated University Hospital of Verona; and Radiology (F.B.P.), Department of Diagnostic and Public Health, Integrated University Hospital of Verona, Italy
| | - Anna Bolzan
- From the Department of Neurosciences (D.M., R.M., A.B., A.I.P., F.C., M.C.), Biomedicine and Movement Sciences, University of Verona; Department of Brain Sciences (R.M.), Imperial College London, Hammersmith Hospital, UK; Department of Oncology and Molecular Medicine (S.R.), Higher Institute of Health Care, Rome; Neuroradiology & Radiology Units (S.M.), Integrated University Hospital of Verona; and Radiology (F.B.P.), Department of Diagnostic and Public Health, Integrated University Hospital of Verona, Italy
| | - Anna Isabella Pisani
- From the Department of Neurosciences (D.M., R.M., A.B., A.I.P., F.C., M.C.), Biomedicine and Movement Sciences, University of Verona; Department of Brain Sciences (R.M.), Imperial College London, Hammersmith Hospital, UK; Department of Oncology and Molecular Medicine (S.R.), Higher Institute of Health Care, Rome; Neuroradiology & Radiology Units (S.M.), Integrated University Hospital of Verona; and Radiology (F.B.P.), Department of Diagnostic and Public Health, Integrated University Hospital of Verona, Italy
| | - Stefania Rossi
- From the Department of Neurosciences (D.M., R.M., A.B., A.I.P., F.C., M.C.), Biomedicine and Movement Sciences, University of Verona; Department of Brain Sciences (R.M.), Imperial College London, Hammersmith Hospital, UK; Department of Oncology and Molecular Medicine (S.R.), Higher Institute of Health Care, Rome; Neuroradiology & Radiology Units (S.M.), Integrated University Hospital of Verona; and Radiology (F.B.P.), Department of Diagnostic and Public Health, Integrated University Hospital of Verona, Italy
| | - Francesco Crescenzo
- From the Department of Neurosciences (D.M., R.M., A.B., A.I.P., F.C., M.C.), Biomedicine and Movement Sciences, University of Verona; Department of Brain Sciences (R.M.), Imperial College London, Hammersmith Hospital, UK; Department of Oncology and Molecular Medicine (S.R.), Higher Institute of Health Care, Rome; Neuroradiology & Radiology Units (S.M.), Integrated University Hospital of Verona; and Radiology (F.B.P.), Department of Diagnostic and Public Health, Integrated University Hospital of Verona, Italy
| | - Stefania Montemezzi
- From the Department of Neurosciences (D.M., R.M., A.B., A.I.P., F.C., M.C.), Biomedicine and Movement Sciences, University of Verona; Department of Brain Sciences (R.M.), Imperial College London, Hammersmith Hospital, UK; Department of Oncology and Molecular Medicine (S.R.), Higher Institute of Health Care, Rome; Neuroradiology & Radiology Units (S.M.), Integrated University Hospital of Verona; and Radiology (F.B.P.), Department of Diagnostic and Public Health, Integrated University Hospital of Verona, Italy
| | - Francesca Benedetta Pizzini
- From the Department of Neurosciences (D.M., R.M., A.B., A.I.P., F.C., M.C.), Biomedicine and Movement Sciences, University of Verona; Department of Brain Sciences (R.M.), Imperial College London, Hammersmith Hospital, UK; Department of Oncology and Molecular Medicine (S.R.), Higher Institute of Health Care, Rome; Neuroradiology & Radiology Units (S.M.), Integrated University Hospital of Verona; and Radiology (F.B.P.), Department of Diagnostic and Public Health, Integrated University Hospital of Verona, Italy
| | - Massimiliano Calabrese
- From the Department of Neurosciences (D.M., R.M., A.B., A.I.P., F.C., M.C.), Biomedicine and Movement Sciences, University of Verona; Department of Brain Sciences (R.M.), Imperial College London, Hammersmith Hospital, UK; Department of Oncology and Molecular Medicine (S.R.), Higher Institute of Health Care, Rome; Neuroradiology & Radiology Units (S.M.), Integrated University Hospital of Verona; and Radiology (F.B.P.), Department of Diagnostic and Public Health, Integrated University Hospital of Verona, Italy.
| |
Collapse
|
22
|
The Role of Dendritic Cells during Physiological and Pathological Dentinogenesis. J Clin Med 2021; 10:jcm10153348. [PMID: 34362130 PMCID: PMC8348392 DOI: 10.3390/jcm10153348] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Revised: 07/25/2021] [Accepted: 07/26/2021] [Indexed: 12/13/2022] Open
Abstract
The dental pulp is a soft connective tissue of ectomesenchymal origin that harbors distinct cell populations, capable of interacting with each other to maintain the vitality of the tooth. After tooth injuries, a sequence of complex biological events takes place in the pulpal tissue to restore its homeostasis. The pulpal response begins with establishing an inflammatory reaction that leads to the formation of a matrix of reactionary or reparative dentin, according to the nature of the exogenous stimuli. Using several in vivo designs, antigen-presenting cells, including macrophages and dendritic cells (DCs), are identified in the pulpal tissue before tertiary dentin deposition under the afflicted area. However, the precise nature of this phenomenon and its relationship to inherent pulp cells are not yet clarified. This literature review aims to discuss the role of pulpal DCs and their relationship to progenitor/stem cells, odontoblasts or odontoblast-like cells, and other immunocompetent cells during physiological and pathological dentinogenesis. The concept of “dentin-pulp immunology” is proposed for understanding the crosstalk among these cell types after tooth injuries, and the possibility of immune-based therapies is introduced to accelerate pulpal healing after exogenous stimuli.
Collapse
|
23
|
Bartonella henselae Persistence within Mesenchymal Stromal Cells Enhances Endothelial Cell Activation and Infectibility That Amplifies the Angiogenic Process. Infect Immun 2021; 89:e0014121. [PMID: 34031126 DOI: 10.1128/iai.00141-21] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Some bacterial pathogens can manipulate the angiogenic response, suppressing or inducing it for their own ends. In humans, Bartonella henselae is associated with cat-scratch disease and vasculoproliferative disorders such as bacillary angiomatosis and bacillary peliosis. Although endothelial cells (ECs) support the pathogenesis of B. henselae, the mechanisms by which B. henselae induces EC activation are not completely clear, as well as the possible contributions of other cells recruited at the site of infection. Mesenchymal stromal cells (MSCs) are endowed with angiogenic potential and play a dual role in infections, exerting antimicrobial properties but also acting as a shelter for pathogens. Here, we delved into the role of MSCs as a reservoir of B. henselae and modulator of EC functions. B. henselae readily infected MSCs and survived in perinuclearly bound vacuoles for up to 8 days. Infection enhanced MSC proliferation and the expression of epidermal growth factor receptor (EGFR), Toll-like receptor 2 (TLR2), and nucleotide-binding oligomerization domain-containing protein 1 (NOD1), proteins that are involved in bacterial internalization and cytokine production. Secretome analysis revealed that infected MSCs secreted higher levels of the proangiogenic factors vascular endothelial growth factor (VEGF), fibroblast growth factor 7 (FGF-7), matrix metallopeptidase 9 (MMP-9), placental growth factor (PIGF), serpin E1, thrombospondin 1 (TSP-1), urokinase-type plasminogen activator (uPA), interleukin 6 (IL-6), platelet-derived growth factor D (PDGF-D), chemokine ligand 5 (CCL5), and C-X-C motif chemokine ligand 8 (CXCL8). Supernatants from B. henselae-infected MSCs increased the susceptibility of ECs to B. henselae infection and enhanced EC proliferation, invasion, and reorganization in tube-like structures. Altogether, these results indicate MSCs as a still underestimated niche for persistent B. henselae infection and reveal MSC-EC cross talk that may contribute to exacerbate bacterium-induced angiogenesis and granuloma formation.
Collapse
|
24
|
Kaleta B. Osteopontin and Transplantation: Where Are We Now? Arch Immunol Ther Exp (Warsz) 2021; 69:15. [PMID: 34019147 PMCID: PMC8139897 DOI: 10.1007/s00005-021-00617-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Accepted: 05/12/2021] [Indexed: 11/26/2022]
Abstract
Organ transplantation represents the optimal therapeutic tool for patients with end-stage organ failure. Hematopoietic stem cell transplantation (HSCT) is likewise an effective therapy for a wide range of malignant and non-malignant diseases. Better understanding of transplantation immunology and the use of multi-modal immunosuppression protocols, can decrease the risk of graft failure and graft-versus-host disease (GVHD) after HSCT. Nevertheless, a major challenge of modern transplantology still seems to be finding non-invasive biomarkers for recipients selection, monitoring of allograft function, and diagnosis of rejection. Since proinflammatory cytokine osteopontin (OPN) is closely involved in regulating both adaptive and innate immune responses, as well as the pathogenesis of inflammatory and autoimmune diseases, it is likely to play an important role in organ and HSC transplantation. This review is to summarize recent advances in our knowledge about OPN function in the kidney, heart, liver, lung, and HSC transplantation. Most studies found that elevated OPN is associated with poorer graft function in kidney, heart, liver and lung recipients. Moreover, some reports suggested that this protein can play role in GVHD pathogenesis. However, due to relatively small number of similar studies, as well as some inconclusive results, future investigation in this field is needed to verify if OPN can serve as a biomarker of organ and HSC transplantation. The knowledge about such markers will promote our understanding of the mechanisms underlying graft dysfunction and posttransplant mortality. In addition, such knowledge may be helpful in the development of new treatment strategies and identification of recipients with increased risk of allograft failure.
Collapse
Affiliation(s)
- Beata Kaleta
- Department of Clinical Immunology, Medical University of Warsaw, Nowogrodzka 59 St., 02-006, Warsaw, Poland.
| |
Collapse
|
25
|
Cappellano G, Vecchio D, Magistrelli L, Clemente N, Raineri D, Barbero Mazzucca C, Virgilio E, Dianzani U, Chiocchetti A, Comi C. The Yin-Yang of osteopontin in nervous system diseases: damage versus repair. Neural Regen Res 2021; 16:1131-1137. [PMID: 33269761 PMCID: PMC8224140 DOI: 10.4103/1673-5374.300328] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Osteopontin is a broadly expressed pleiotropic protein, and is attracting increased attention because of its role in the pathophysiology of several inflammatory, degenerative, autoimmune, and oncologic diseases. In fact, in the last decade, several studies have shown that osteopontin contributes to tissue damage not only by recruiting harmful inflammatory cells to the site of lesion, but also increasing their survival. The detrimental role of osteopontin has been indeed well documented in the context of different neurological conditions (i.e., multiple sclerosis, Parkinson's, and Alzheimer's diseases). Intriguingly, recent findings show that osteopontin is involved not only in promoting tissue damage (the Yin), but also in repair/regenerative mechanisms (the Yang), mostly triggered by the inflammatory response. These two apparently discordant roles are partly related to the presence of different functional domains in the osteopontin molecule, which are exposed after thrombin or metalloproteases cleavages. Such functional domains may in turn activate intracellular signaling pathways and mediate cell-cell and cell-matrix interactions. This review describes the current knowledge on the Yin and Yang features of osteopontin in nervous system diseases. Understanding the mechanisms behind the Yin/Yang would be relevant to develop highly specific tools targeting this multifunctional protein.
Collapse
Affiliation(s)
- Giuseppe Cappellano
- Department of Health Sciences, Interdisciplinary Research Center of Autoimmune Diseases (IRCAD); Center for Translational Research on Autoimmune and Allergic Disease-CAAD, University of Piemonte Orientale, Novara, Italy
| | - Domizia Vecchio
- Department of Translational Medicine, Neurology Unit, Interdisciplinary Research Center of Autoimmune Diseases (IRCAD), University of Piemonte Orientale, Novara, Italy
| | - Luca Magistrelli
- Department of Translational Medicine, Neurology Unit, Interdisciplinary Research Center of Autoimmune Diseases (IRCAD), University of Piemonte Orientale, Novara; PhD Program in Clinical and Experimental Medicine and Medical Humanities, University of Insubria, Varese, Italy
| | - Nausicaa Clemente
- Department of Health Sciences, Interdisciplinary Research Center of Autoimmune Diseases (IRCAD), University of Piemonte Orientale, Novara, Italy
| | - Davide Raineri
- Department of Health Sciences, Interdisciplinary Research Center of Autoimmune Diseases (IRCAD); Center for Translational Research on Autoimmune and Allergic Disease-CAAD, University of Piemonte Orientale, Novara, Italy
| | - Camilla Barbero Mazzucca
- Department of Health Sciences, Interdisciplinary Research Center of Autoimmune Diseases (IRCAD); Center for Translational Research on Autoimmune and Allergic Disease-CAAD, University of Piemonte Orientale, Novara, Italy
| | - Eleonora Virgilio
- Department of Translational Medicine, Neurology Unit, Interdisciplinary Research Center of Autoimmune Diseases (IRCAD), University of Piemonte Orientale, Novara, Italy
| | - Umberto Dianzani
- Department of Health Sciences, Interdisciplinary Research Center of Autoimmune Diseases (IRCAD); Center for Translational Research on Autoimmune and Allergic Disease-CAAD, University of Piemonte Orientale, Novara, Italy
| | - Annalisa Chiocchetti
- Department of Health Sciences, Interdisciplinary Research Center of Autoimmune Diseases (IRCAD); Center for Translational Research on Autoimmune and Allergic Disease-CAAD, University of Piemonte Orientale, Novara, Italy
| | - Cristoforo Comi
- Department of Translational Medicine, Neurology Unit, Interdisciplinary Research Center of Autoimmune Diseases (IRCAD), University of Piemonte Orientale, Novara, Italy
| |
Collapse
|
26
|
The bovine Lactoferrin-Osteopontin complex increases proliferation of human intestinal epithelial cells by activating the PI3K/Akt signaling pathway. Food Chem 2020; 310:125919. [DOI: 10.1016/j.foodchem.2019.125919] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2019] [Revised: 11/06/2019] [Accepted: 11/16/2019] [Indexed: 12/17/2022]
|
27
|
Costantini C, Bellet MM, Pariano M, Renga G, Stincardini C, Goldstein AL, Garaci E, Romani L. A Reappraisal of Thymosin Alpha1 in Cancer Therapy. Front Oncol 2019; 9:873. [PMID: 31555601 PMCID: PMC6742685 DOI: 10.3389/fonc.2019.00873] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2019] [Accepted: 08/22/2019] [Indexed: 02/06/2023] Open
Abstract
Thymosin alpha1 (Tα1), an endogenous peptide first isolated from the thymic tissue in the mid-sixties, has gained considerable attention for its immunostimulatory activity that led to its application to diverse pathological conditions, including cancer. Studies in animal models and human patients have shown promising results in different types of malignancies, especially when Tα1 was used in combination with other chemo- and immune therapies. For this reason, the advancements in our knowledge on the adjuvant role of Tα1 have moved in parallel with the development of novel cancer therapies in a way that Tα1 was integrated to changing paradigms and protocols, and tested for increased efficacy and safety. Cancer immunotherapy has recently experienced a tremendous boost following the development and clinical application of immune checkpoint inhibitors. By unleashing the full potential of the adaptive immune response, checkpoint inhibitors were expected to be very effective against tumors, but it soon became clear that a widespread and successful application was not straightforward and shortcomings in efficacy and safety clearly emerged. This scenario led to the development of novel concepts in immunotherapy and the design of combination protocols to overcome these limitations, thus opening up novel opportunities for Tα1 application. Herein, we summarize in a historical perspective the use of Tα1 in cancer, with particular reference to melanoma, hepatocellular carcinoma and lung cancer. We will discuss the current limitations of checkpoint inhibitors in clinical practice and the mechanisms at the basis of a potential application of Tα1 in combination protocols.
Collapse
Affiliation(s)
- Claudio Costantini
- Department of Experimental Medicine, University of Perugia, Perugia, Italy
| | - Marina M Bellet
- Department of Experimental Medicine, University of Perugia, Perugia, Italy
| | - Marilena Pariano
- Department of Experimental Medicine, University of Perugia, Perugia, Italy
| | - Giorgia Renga
- Department of Experimental Medicine, University of Perugia, Perugia, Italy
| | | | - Allan L Goldstein
- Department of Biochemistry and Molecular Medicine, School of Medicine and Health Sciences, The George Washington University, Washington, DC, United States
| | - Enrico Garaci
- University San Raffaele and IRCCS San Raffaele, Rome, Italy
| | - Luigina Romani
- Department of Experimental Medicine, University of Perugia, Perugia, Italy
| |
Collapse
|