1
|
Matsuguma C, Takahashi K, Okada S, Tokitaka R, Hamano H, Kaneyasu H, Fujimoto Y, Hasegawa S. Clinical utility of gastric fluid cytokine levels in preterm infants for predicting histological chorioamnionitis. Cytokine 2024; 180:156642. [PMID: 38749278 DOI: 10.1016/j.cyto.2024.156642] [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: 02/16/2024] [Revised: 04/25/2024] [Accepted: 05/10/2024] [Indexed: 06/21/2024]
Abstract
BACKGROUND The risk of various complications, such as neonatal death, early onset sepsis, and chronic lung disease, is increased in infants born to mothers with chorioamnionitis (CAM). However, predicting the diagnosis of histological CAM (hCAM) in the early postnatal period is challenging for clinicians due to pathological considerations. Therefore, an early diagnostic tool for hCAM is needed. Gastric fluid at birth is considered a suitable biomarker for predicting the intrauterine environment because most of its components are from amniotic fluid, and the sampling technique is less invasive. This study aimed to evaluate the clinical utility of cytokines in the gastric fluid of preterm infants at birth as predictors of hCAM. METHODS We retrieved gastric fluid and serum from 21 preterm infants with a gestational age of ≤ 32 weeks within 1 h after birth and used cytometric bead array to measure the concentrations of interleukin (IL)-2, IL-4, IL-6, IL-10, tumor necrosis factor-alpha, and interferon-gamma. We compared the cytokine concentrations in the gastric fluid and serum of the preterm infants born to mothers with or without hCAM. RESULTS The gastric fluid, serum IL-6, and serum IL-10 concentrations were significantly higher in the hCAM group than that in the non-hCAM group. The best cutoff values for predicting hCAM was > 2,855 pg/mL and > 315 pg/mL for IL-6 in the gastric fluid and serum, respectively. Receiver operating characteristic curves showed that gastric fluid IL-6 concentrations correlated more strongly with the presence of hCAM than serum IL-6 concentrations. CONCLUSION IL-6 in the gastric fluid at birth may be a more promising biomarker for predicting the presence of hCAM than that in serum. IL-6 concentration analysis in the gastric fluid at birth might help to diagnose hCAM immediately after birth and improve the prognosis of preterm infants.
Collapse
Affiliation(s)
- Chie Matsuguma
- Department of Pediatrics, Yamaguchi University Graduate School of Medicine, Ube, Japan
| | - Kazumasa Takahashi
- Department of Pediatrics, Yamaguchi University Graduate School of Medicine, Ube, Japan.
| | - Seigo Okada
- Department of Pediatrics, Yamaguchi University Graduate School of Medicine, Ube, Japan
| | - Rui Tokitaka
- Department of Pediatrics, Yamaguchi University Graduate School of Medicine, Ube, Japan
| | - Hiroki Hamano
- Department of Pediatrics, Yamaguchi University Graduate School of Medicine, Ube, Japan
| | - Hidenobu Kaneyasu
- Department of Pediatrics, Yamaguchi University Graduate School of Medicine, Ube, Japan
| | - Yousuke Fujimoto
- Department of Pediatrics, Yamaguchi University Graduate School of Medicine, Ube, Japan
| | - Shunji Hasegawa
- Department of Pediatrics, Yamaguchi University Graduate School of Medicine, Ube, Japan
| |
Collapse
|
2
|
Xiao R, Lin M, Liu M, Ma Q. Single cells and TRUST4 reveal immunological features of the HFRS transcriptome. Front Med (Lausanne) 2024; 11:1403335. [PMID: 38803345 PMCID: PMC11128564 DOI: 10.3389/fmed.2024.1403335] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Accepted: 04/24/2024] [Indexed: 05/29/2024] Open
Abstract
The etiology of hemorrhagic fever with renal syndrome (HFRS) is significantly impacted by a variety of immune cells. Nevertheless, the existing techniques for sequencing peripheral blood T cell receptor (TCR) or B cell receptor (BCR) libraries in HFRS are constrained by both limitations and high costs. In this investigation, we utilized the computational tool TRUST4 to generate TCR and BCR libraries utilizing comprehensive RNA-seq data from peripheral blood specimens of HFRS patients. This facilitated the examination of clonality and diversity within immune libraries linked to the condition. Despite previous research on immune cell function, the underlying mechanisms remain intricate, and differential gene expression across immune cell types and cell-to-cell interactions within immune cell clusters have not been thoroughly explored. To address this gap, we performed clustering analysis on 11 cell subsets derived from raw single-cell RNA-seq data, elucidating characteristic changes in cell subset proportions under disease conditions. Additionally, we utilized CellChat, a tool for cell-cell communication analysis, to investigate the impact of MIF family, CD70 family, and GALECTIN family cytokines-known to be involved in cell communication-on immune cell subsets. Furthermore, hdWGCNA analysis identified core genes implicated in HFRS pathogenesis within T cells and B cells. Trajectory analysis revealed that most cell subsets were in a developmental stage, with high expression of transcription factors such as NFKB and JUN in Effector CD8+ T cells, as well as in Naive CD4+ T cells and Naive B cells. Our findings provide a comprehensive understanding of the dynamic changes in immune cells during HFRS pathogenesis, identifying specific V genes and J genes in TCR and BCR that contribute to advancing our knowledge of HFRS. These insights offer potential implications for the diagnosis and treatment of this autoimmune disease.
Collapse
Affiliation(s)
| | | | | | - Qingqing Ma
- The Central Laboratory of Guizhou Aerospace Hospital, Zunyi, China
| |
Collapse
|
3
|
Tan AW, Tong X, Alvarez-Cubela S, Chen P, Santana AG, Morales AA, Tian R, Infante R, Nunes de Paiva V, Kulandavelu S, Benny M, Dominguez-Bendala J, Wu S, Young KC, Rodrigues CO, Schmidt AF. c-Myc Drives inflammation of the maternal-fetal interface, and neonatal lung remodeling induced by intra-amniotic inflammation. Front Cell Dev Biol 2024; 11:1245747. [PMID: 38481391 PMCID: PMC10933046 DOI: 10.3389/fcell.2023.1245747] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2023] [Accepted: 12/07/2023] [Indexed: 04/11/2024] Open
Abstract
Background: Intra-amniotic inflammation (IAI) is associated with increased risk of preterm birth and bronchopulmonary dysplasia (BPD), but the mechanisms by which IAI leads to preterm birth and BPD are poorly understood, and there are no effective therapies for preterm birth and BPD. The transcription factor c-Myc regulates various biological processes like cell growth, apoptosis, and inflammation. We hypothesized that c-Myc modulates inflammation at the maternal-fetal interface, and neonatal lung remodeling. The objectives of our study were 1) to determine the kinetics of c-Myc in the placenta, fetal membranes and neonatal lungs exposed to IAI, and 2) to determine the role of c-Myc in modulating inflammation at the maternal-fetal interface, and neonatal lung remodeling induced by IAI. Methods: Pregnant Sprague-Dawley rats were randomized into three groups: 1) Intra-amniotic saline injections only (control), 2) Intra-amniotic lipopolysaccharide (LPS) injections only, and 3) Intra-amniotic LPS injections with c-Myc inhibitor 10058-F4. c-Myc expression, markers of inflammation, angiogenesis, immunohistochemistry, and transcriptomic analyses were performed on placenta and fetal membranes, and neonatal lungs to determine kinetics of c-Myc expression in response to IAI, and effects of prenatal systemic c-Myc inhibition on lung remodeling at postnatal day 14. Results: c-Myc was upregulated in the placenta, fetal membranes, and neonatal lungs exposed to IAI. IAI caused neutrophil infiltration and neutrophil extracellular trap (NET) formation in the placenta and fetal membranes, and neonatal lung remodeling with pulmonary hypertension consistent with a BPD phenotype. Prenatal inhibition of c-Myc with 10058-F4 in IAI decreased neutrophil infiltration and NET formation, and improved neonatal lung remodeling induced by LPS, with improved alveolarization, increased angiogenesis, and decreased pulmonary vascular remodeling. Discussion: In a rat model of IAI, c-Myc regulates neutrophil recruitment and NET formation in the placenta and fetal membranes. c-Myc also participates in neonatal lung remodeling induced by IAI. Further studies are needed to investigate c-Myc as a potential therapeutic target for IAI and IAI-associated BPD.
Collapse
Affiliation(s)
- April W. Tan
- Division of Neonatology, Department of Pediatrics, University of Miami Miller School of Medicine/Holtz Children’s Hospital, Miami, FL, United States
| | - Xiaoying Tong
- Division of Neonatology, Department of Pediatrics, University of Miami Miller School of Medicine/Holtz Children’s Hospital, Miami, FL, United States
| | - Silvia Alvarez-Cubela
- Diabetes Research Institute, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Pingping Chen
- Division of Neonatology, Department of Pediatrics, University of Miami Miller School of Medicine/Holtz Children’s Hospital, Miami, FL, United States
| | - Aline Guimarães Santana
- Department of Biomedical Science, Florida Atlantic University Charles E. Schmidt College of Medicine, Boca Raton, FL, United States
| | - Alejo A. Morales
- Department of Molecular and Cellular Pharmacology, University of Miami Leonard M. Miller School of Medicine, Miami, FL, United States
| | - Runxia Tian
- Division of Neonatology, Department of Pediatrics, University of Miami Miller School of Medicine/Holtz Children’s Hospital, Miami, FL, United States
| | - Rae Infante
- Division of Neonatology, Department of Pediatrics, University of Miami Miller School of Medicine/Holtz Children’s Hospital, Miami, FL, United States
| | - Vanessa Nunes de Paiva
- Division of Neonatology, Department of Pediatrics, University of Miami Miller School of Medicine/Holtz Children’s Hospital, Miami, FL, United States
| | - Shathiyah Kulandavelu
- Division of Pediatric Nephrology, Department of Pediatrics, University of Miami Miller School of Medicine, Miami, FL, United States
- Interdisciplinary Stem Cell Institute, University of Miami Leonard M. Miller School of Medicine, Miami, FL, United States
| | - Merline Benny
- Division of Neonatology, Department of Pediatrics, University of Miami Miller School of Medicine/Holtz Children’s Hospital, Miami, FL, United States
| | - Juan Dominguez-Bendala
- Diabetes Research Institute, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Shu Wu
- Division of Neonatology, Department of Pediatrics, University of Miami Miller School of Medicine/Holtz Children’s Hospital, Miami, FL, United States
| | - Karen C. Young
- Division of Neonatology, Department of Pediatrics, University of Miami Miller School of Medicine/Holtz Children’s Hospital, Miami, FL, United States
| | - Claudia O. Rodrigues
- Department of Biomedical Science, Florida Atlantic University Charles E. Schmidt College of Medicine, Boca Raton, FL, United States
- Department of Molecular and Cellular Pharmacology, University of Miami Leonard M. Miller School of Medicine, Miami, FL, United States
- Interdisciplinary Stem Cell Institute, University of Miami Leonard M. Miller School of Medicine, Miami, FL, United States
| | - Augusto F. Schmidt
- Division of Neonatology, Department of Pediatrics, University of Miami Miller School of Medicine/Holtz Children’s Hospital, Miami, FL, United States
| |
Collapse
|
4
|
Luo Z, Wei Z, Zhang G, Chen H, Li L, Kang X. Achilles' Heel-The Significance of Maintaining Microenvironmental Homeostasis in the Nucleus Pulposus for Intervertebral Discs. Int J Mol Sci 2023; 24:16592. [PMID: 38068915 PMCID: PMC10706299 DOI: 10.3390/ijms242316592] [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: 09/15/2023] [Revised: 11/02/2023] [Accepted: 11/07/2023] [Indexed: 12/18/2023] Open
Abstract
The dysregulation of intracellular and extracellular environments as well as the aberrant expression of ion channels on the cell membrane are intricately linked to a diverse array of degenerative disorders, including intervertebral disc degeneration. This condition is a significant contributor to low back pain, which poses a substantial burden on both personal quality of life and societal economics. Changes in the number and function of ion channels can disrupt the water and ion balance both inside and outside cells, thereby impacting the physiological functions of tissues and organs. Therefore, maintaining ion homeostasis and stable expression of ion channels within the cellular microenvironment may prove beneficial in the treatment of disc degeneration. Aquaporin (AQP), calcium ion channels, and acid-sensitive ion channels (ASIC) play crucial roles in regulating water, calcium ions, and hydrogen ions levels. These channels have significant effects on physiological and pathological processes such as cellular aging, inflammatory response, stromal decomposition, endoplasmic reticulum stress, and accumulation of cell metabolites. Additionally, Piezo 1, transient receptor potential vanilloid type 4 (TRPV4), tension response enhancer binding protein (TonEBP), potassium ions, zinc ions, and tungsten all play a role in the process of intervertebral disc degeneration. This review endeavors to elucidate alterations in the microenvironment of the nucleus pulposus during intervertebral disc degeneration (IVDD), with a view to offer novel insights and approaches for exploring therapeutic interventions against disc degeneration.
Collapse
Affiliation(s)
- Zhangbin Luo
- Department of Orthopedics, Lanzhou University Second Hospital, Lanzhou 730030, China; (Z.L.); (Z.W.); (G.Z.); (H.C.); (L.L.)
- The Second Clinical Medical College, Lanzhou University, Lanzhou 730030, China
| | - Ziyan Wei
- Department of Orthopedics, Lanzhou University Second Hospital, Lanzhou 730030, China; (Z.L.); (Z.W.); (G.Z.); (H.C.); (L.L.)
- The Second Clinical Medical College, Lanzhou University, Lanzhou 730030, China
| | - Guangzhi Zhang
- Department of Orthopedics, Lanzhou University Second Hospital, Lanzhou 730030, China; (Z.L.); (Z.W.); (G.Z.); (H.C.); (L.L.)
- The Second Clinical Medical College, Lanzhou University, Lanzhou 730030, China
| | - Haiwei Chen
- Department of Orthopedics, Lanzhou University Second Hospital, Lanzhou 730030, China; (Z.L.); (Z.W.); (G.Z.); (H.C.); (L.L.)
| | - Lei Li
- Department of Orthopedics, Lanzhou University Second Hospital, Lanzhou 730030, China; (Z.L.); (Z.W.); (G.Z.); (H.C.); (L.L.)
- The Second Clinical Medical College, Lanzhou University, Lanzhou 730030, China
| | - Xuewen Kang
- Department of Orthopedics, Lanzhou University Second Hospital, Lanzhou 730030, China; (Z.L.); (Z.W.); (G.Z.); (H.C.); (L.L.)
- The Second Clinical Medical College, Lanzhou University, Lanzhou 730030, China
- Key Laboratory of Orthopedics Disease of Gansu Province, Lanzhou University Second Hospital, Lanzhou 730030, China
| |
Collapse
|
5
|
Buchanan CQ, Lawlor ML, Okafor C, Kurian SR, Philip AE, Finkle AE, McQuillan JJ, Haridas S, Koenig JM. Linked Th17 and Calgranulin Responses in Maternal-cord Blood Dyads of Preterm Gestations with Histologic Chorioamnionitis. NEWBORN (CLARKSVILLE, MD.) 2023; 2:133-141. [PMID: 37790838 PMCID: PMC10547109 DOI: 10.5005/jp-journals-11002-0064] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 10/05/2023]
Abstract
Introduction Maternal-fetal immune crosstalk mechanisms are increasingly identified in the pathogenesis of gestational disorders, including histologic chorioamnionitis (HCA). Although an inflammatory Th17 immune phenotype has been described in preterm neonates with HCA, the associated maternal Th17 response is relatively unknown. To refine our understanding of Th17 biology in this context, we examined Th17 responses in maternal-cord blood dyads of preterm gestations. Materials and methods Paired maternal and cord blood (CB) samples were prospectively collected from preterm gestations (23-34 weeks) with HCA or controls. Th17-linked cell frequencies and plasma calgranulin (S100A8, S100A12) levels were determined by flow cytometry and enzyme-linked immunoassay, respectively. Results Analyses of 47 maternal-cord blood pairs showed striking parallel increases in Th17 cell frequencies as well as plasma calgranulin levels in the presence of fetal inflammation. Cord blood S100A12 levels were directly correlated with Th17 cell frequencies. In CB cultures, rh-S100A12 promoted in vitro propagation of Th17-type CD4+ cells. Conclusions Maternal and CB Th17-linked responses are dually amplified in gestations with HCA, supporting a biological role for maternal-fetal interactions in this disorder. In addition to advancing current knowledge of neonatal Th17 mechanisms, these data shed new light on their association with maternal inflammation.
Collapse
Affiliation(s)
- Christopher Q Buchanan
- Department of Obstetrics, Gynecology and Women’s Health, Division of Maternal–Fetal Medicine, Saint Louis University School of Medicine, St. Louis, Missouri, United States of America
| | - Megan L Lawlor
- Department of Obstetrics, Gynecology and Women’s Health, Division of Maternal–Fetal Medicine, Saint Louis University School of Medicine, St. Louis, Missouri, United States of America
| | - Chukwuebuka Okafor
- Department of Pediatrics, Division of Neonatal–Perinatal Medicine, Saint Louis University School of Medicine, St. Louis, Missouri, United States of America
| | - Shannon R Kurian
- Department of Pediatrics, Division of Neonatal–Perinatal Medicine, Saint Louis University School of Medicine, St. Louis, Missouri, United States of America
| | - Andrea E Philip
- Department of Pediatrics, Division of Neonatal–Perinatal Medicine, Saint Louis University School of Medicine, St. Louis, Missouri, United States of America
| | - Abigael E Finkle
- Department of Pediatrics, Division of Neonatal–Perinatal Medicine, Saint Louis University School of Medicine, St. Louis, Missouri, United States of America
| | - Jay J McQuillan
- Department of Pediatrics, Division of Neonatal–Perinatal Medicine, Saint Louis University School of Medicine, St. Louis, Missouri, United States of America
| | - Seema Haridas
- Department of Pediatrics, Division of Neonatal–Perinatal Medicine, Saint Louis University School of Medicine, St. Louis, Missouri, United States of America
| | - Joyce M Koenig
- Department of Pediatrics, Division of Neonatal–Perinatal Medicine, Saint Louis University School of Medicine, St. Louis, Missouri, United States of America
- Department of Molecular Microbiology and Immunology, Saint Louis University School of Medicine, St. Louis, Missouri, United States of America
| |
Collapse
|
6
|
Singh P, Ali SA. Multifunctional Role of S100 Protein Family in the Immune System: An Update. Cells 2022; 11:cells11152274. [PMID: 35892571 PMCID: PMC9332480 DOI: 10.3390/cells11152274] [Citation(s) in RCA: 62] [Impact Index Per Article: 31.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Revised: 07/12/2022] [Accepted: 07/21/2022] [Indexed: 12/04/2022] Open
Abstract
S100 is a broad subfamily of low-molecular weight calcium-binding proteins (9–14 kDa) with structural similarity and functional discrepancy. It is required for inflammation and cellular homeostasis, and can work extracellularly, intracellularly, or both. S100 members participate in a variety of activities in a healthy cell, including calcium storage and transport (calcium homeostasis). S100 isoforms that have previously been shown to play important roles in the immune system as alarmins (DAMPs), antimicrobial peptides, pro-inflammation stimulators, chemo-attractants, and metal scavengers during an innate immune response. Currently, during the pandemic, it was found that several members of the S100 family are implicated in the pathophysiology of COVID-19. Further, S100 family protein members were proposed to be used as a prognostic marker for COVID-19 infection identification using a nasal swab. In the present review, we compiled the vast majority of recent studies that focused on the multifunctionality of S100 proteins in the complex immune system and its associated activities. Furthermore, we shed light on the numerous molecular approaches and signaling cascades regulated by S100 proteins during immune response. In addition, we discussed the involvement of S100 protein members in abnormal defense systems during the pathogenesis of COVID-19.
Collapse
Affiliation(s)
- Parul Singh
- Cell Biology and Proteomics Lab, Animal Biotechnology Center, ICAR-NDRI, Karnal 132001, India;
| | - Syed Azmal Ali
- Cell Biology and Proteomics Lab, Animal Biotechnology Center, ICAR-NDRI, Karnal 132001, India;
- Division of Proteomics of Stem Cells and Cancer, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
- Correspondence: ; Tel.: +91-8708591790
| |
Collapse
|
7
|
Matute JD, Finander B, Pepin D, Ai X, Smith NP, Li JZ, Edlow AG, Villani AC, Lerou PH, Kalish BT. Single-cell immunophenotyping of the fetal immune response to maternal SARS-CoV-2 infection in late gestation. Pediatr Res 2022; 91:1090-1098. [PMID: 34750520 PMCID: PMC8573077 DOI: 10.1038/s41390-021-01793-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Revised: 08/31/2021] [Accepted: 09/28/2021] [Indexed: 02/01/2023]
Abstract
BACKGROUND During the COVID-19 pandemic, thousands of pregnant women have been infected with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). The implications of maternal SARS-CoV-2 infection on fetal and childhood well-being need to be characterized. We aimed to characterize the fetal immune response to maternal SARS-CoV-2 infection. METHODS We performed single-cell RNA-sequencing and T cell receptor sequencing on cord blood mononuclear cells (CBMCs) from newborns of mothers infected with SARS-CoV-2 in the third trimester (cases) or without SARS-CoV-2 infection (controls). RESULTS We identified widespread gene expression changes in CBMCs from cases, including upregulation of interferon-stimulated genes and major histocompatibility complex genes in CD14+ monocytes, transcriptional changes suggestive of activation of plasmacytoid dendritic cells, and activation and exhaustion of natural killer cells. Lastly, we observed fetal T cell clonal expansion in cases compared to controls. CONCLUSIONS As none of the infants were infected with SARS-CoV-2, our results suggest that maternal SARS-CoV-2 infection might modulate the fetal immune system in the absence of vertical transmission. IMPACT The implications of maternal SARS-CoV-2 infection in the absence of vertical transmission on fetal and childhood well-being are poorly understood. Maternal SARS-CoV-2 infection might modulate the fetal immune system in the absence of vertical transmission. This study raises important questions about the untoward effects of maternal SARS-CoV-2 on the fetus, even in the absence of vertical transmission.
Collapse
Affiliation(s)
- Juan D Matute
- Division of Neonatology and Newborn Medicine, Department of Pediatrics, Massachusetts General Hospital (MGH), Boston, MA, USA.
| | - Benjamin Finander
- Division of Newborn Medicine, Department of Pediatrics, Boston Children's Hospital, Boston, MA, USA
| | - David Pepin
- Department of Pediatric Surgery, MGH, Boston, MA, USA
| | - Xingbin Ai
- Division of Neonatology and Newborn Medicine, Department of Pediatrics, Massachusetts General Hospital (MGH), Boston, MA, USA
| | | | - Jonathan Z Li
- Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - Andrea G Edlow
- Department of Obstetrics and Gynecology, MGH, Boston, MA, USA
| | | | - Paul H Lerou
- Division of Neonatology and Newborn Medicine, Department of Pediatrics, Massachusetts General Hospital (MGH), Boston, MA, USA
| | - Brian T Kalish
- Division of Newborn Medicine, Department of Pediatrics, Boston Children's Hospital, Boston, MA, USA.
- Division of Neonatology, Department of Paediatrics, The Hospital for Sick Children, Toronto, ON, Canada.
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
8
|
Abdelfattah N, Kumar P, Wang C, Leu JS, Flynn WF, Gao R, Baskin DS, Pichumani K, Ijare OB, Wood SL, Powell SZ, Haviland DL, Parker Kerrigan BC, Lang FF, Prabhu SS, Huntoon KM, Jiang W, Kim BYS, George J, Yun K. Single-cell analysis of human glioma and immune cells identifies S100A4 as an immunotherapy target. Nat Commun 2022; 13:767. [PMID: 35140215 PMCID: PMC8828877 DOI: 10.1038/s41467-022-28372-y] [Citation(s) in RCA: 175] [Impact Index Per Article: 87.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Accepted: 01/17/2022] [Indexed: 12/24/2022] Open
Abstract
A major rate-limiting step in developing more effective immunotherapies for GBM is our inadequate understanding of the cellular complexity and the molecular heterogeneity of immune infiltrates in gliomas. Here, we report an integrated analysis of 201,986 human glioma, immune, and other stromal cells at the single cell level. In doing so, we discover extensive spatial and molecular heterogeneity in immune infiltrates. We identify molecular signatures for nine distinct myeloid cell subtypes, of which five are independent prognostic indicators of glioma patient survival. Furthermore, we identify S100A4 as a regulator of immune suppressive T and myeloid cells in GBM and demonstrate that deleting S100a4 in non-cancer cells is sufficient to reprogram the immune landscape and significantly improve survival. This study provides insights into spatial, molecular, and functional heterogeneity of glioma and glioma-associated immune cells and demonstrates the utility of this dataset for discovering therapeutic targets for this poorly immunogenic cancer.
Collapse
Affiliation(s)
- Nourhan Abdelfattah
- Department of Neurology, Houston Methodist Research Institute, Houston, TX, USA
| | - Parveen Kumar
- The Jackson Laboratory for Genomic Medicine, Farmington, CT, USA
| | - Caiyi Wang
- Department of Neurology, Houston Methodist Research Institute, Houston, TX, USA
- Xiangya Hospital, Central South University, Changsha, P. R. China
| | - Jia-Shiun Leu
- Department of Neurology, Houston Methodist Research Institute, Houston, TX, USA
| | - William F Flynn
- The Jackson Laboratory for Genomic Medicine, Farmington, CT, USA
| | - Ruli Gao
- Center for Bioinformatics and Computational Biology. Houston Methodist Research Institute Houston, Houston, TX, USA
| | - David S Baskin
- Department of Neurosurgery, Houston Methodist Neurological Institute, Houston, TX, USA
- Kenneth R. Peak Center for Brain and Pituitary Tumor Treatment and Research, Department of Neurosurgery, Houston Methodist Neurological Institute, Houston, TX, USA
- Department of Neurosurgery, Weill Cornell Medical College, New York, NY, USA
| | - Kumar Pichumani
- Department of Neurosurgery, Houston Methodist Neurological Institute, Houston, TX, USA
- Kenneth R. Peak Center for Brain and Pituitary Tumor Treatment and Research, Department of Neurosurgery, Houston Methodist Neurological Institute, Houston, TX, USA
- Department of Neurosurgery, Weill Cornell Medical College, New York, NY, USA
| | - Omkar B Ijare
- Department of Neurosurgery, Houston Methodist Neurological Institute, Houston, TX, USA
- Kenneth R. Peak Center for Brain and Pituitary Tumor Treatment and Research, Department of Neurosurgery, Houston Methodist Neurological Institute, Houston, TX, USA
| | - Stephanie L Wood
- Department of Neurosurgery, Houston Methodist Neurological Institute, Houston, TX, USA
| | - Suzanne Z Powell
- Kenneth R. Peak Center for Brain and Pituitary Tumor Treatment and Research, Department of Neurosurgery, Houston Methodist Neurological Institute, Houston, TX, USA
- Department of Neurosurgery, Weill Cornell Medical College, New York, NY, USA
- Department of Pathology and Genomic Medicine, Houston Methodist Hospital, Houston, TX, USA
- Department of Pathology and Laboratory Medicine, Weill Cornell Medical College, New York, NY, USA
| | - David L Haviland
- Flow Cytometry Core, Houston Methodist Research Institute, Houston, TX, USA
| | - Brittany C Parker Kerrigan
- Department of Neurosurgery, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- The Brain Tumor Center, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd., Houston, TX, USA
| | - Frederick F Lang
- Department of Neurosurgery, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- The Brain Tumor Center, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd., Houston, TX, USA
| | - Sujit S Prabhu
- Department of Neurosurgery, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Kristin M Huntoon
- Department of Neurosurgery, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- The Brain Tumor Center, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd., Houston, TX, USA
| | - Wen Jiang
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Betty Y S Kim
- Department of Neurosurgery, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- The Brain Tumor Center, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd., Houston, TX, USA
| | - Joshy George
- The Jackson Laboratory for Genomic Medicine, Farmington, CT, USA
| | - Kyuson Yun
- Department of Neurology, Houston Methodist Research Institute, Houston, TX, USA.
- Department of Neurology, Weill Cornell Medical College, New York, NY, USA.
| |
Collapse
|
9
|
Motomura K, Romero R, Plazyo O, Garcia-Flores V, Gershater M, Galaz J, Miller D, Gomez-Lopez N. The alarmin S100A12 causes sterile inflammation of the human chorioamniotic membranes as well as preterm birth and neonatal mortality in mice†. Biol Reprod 2021; 105:1494-1509. [PMID: 34632484 PMCID: PMC8689293 DOI: 10.1093/biolre/ioab188] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Revised: 09/23/2021] [Accepted: 10/06/2021] [Indexed: 01/08/2023] Open
Abstract
Sterile inflammation is triggered by danger signals, or alarmins, released upon cellular stress or necrosis. Sterile inflammation occurring in the amniotic cavity (i.e. sterile intra-amniotic inflammation) is frequently observed in women with spontaneous preterm labor resulting in preterm birth, the leading cause of neonatal morbidity and mortality worldwide; this condition is associated with increased amniotic fluid concentrations of alarmins. However, the mechanisms whereby alarmins induce sterile intra-amniotic inflammation are still under investigation. Herein, we investigated the mechanisms whereby the alarmin S100A12 induces inflammation of the human chorioamniotic membranes in vitro and used a mouse model to establish a causal link between this alarmin and adverse perinatal outcomes. We report that S100A12 initiates sterile inflammation in the chorioamniotic membranes by upregulating the expression of inflammatory mediators such as pro-inflammatory cytokines and pattern recognition receptors. Importantly, S100A12 induced the priming and activation of inflammasomes, resulting in caspase-1 cleavage and the subsequent release of mature IL-1β by the chorioamniotic membranes. This alarmin also caused the activation of the chorioamniotic membranes by promoting MMP-2 activity and collagen degradation. Lastly, the ultrasound-guided intra-amniotic injection of S100A12 at specific concentrations observed in the majority of women with sterile intra-amniotic inflammation induced preterm birth (rates: 17% at 200 ng/sac; 25% at 300 ng/sac; 25% at 400 ng/sac) and neonatal mortality (rates: 22% at 200 ng/sac; 44% at 300 ng/sac; 31% at 400 ng/sac), thus demonstrating a causal link between this alarmin and adverse perinatal outcomes. Collectively, our findings shed light on the inflammatory responses driven by alarmins in the chorioamniotic membranes, providing insight into the immune mechanisms leading to preterm birth in women with sterile intra-amniotic inflammation.
Collapse
Affiliation(s)
- Kenichiro Motomura
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, US Department of Health and Human Services (NICHD/NIH/DHHS), Bethesda, Maryland, and Detroit, Michigan, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, Michigan, USA
| | - Roberto Romero
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, US Department of Health and Human Services (NICHD/NIH/DHHS), Bethesda, Maryland, and Detroit, Michigan, USA
- Department of Obstetrics and Gynecology, University of Michigan, Ann Arbor, Michigan, USA
- Department of Epidemiology and Biostatistics, Michigan State University, East Lansing, Michigan, USA
- Center for Molecular Medicine and Genetics, Wayne State University, Detroit, Michigan, USA
- Detroit Medical Center, Detroit, Michigan, USA
| | - Olesya Plazyo
- Department of Dermatology, University of Michigan, Ann Arbor, Michigan, USA
| | - Valeria Garcia-Flores
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, US Department of Health and Human Services (NICHD/NIH/DHHS), Bethesda, Maryland, and Detroit, Michigan, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, Michigan, USA
| | - Meyer Gershater
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, US Department of Health and Human Services (NICHD/NIH/DHHS), Bethesda, Maryland, and Detroit, Michigan, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, Michigan, USA
| | - Jose Galaz
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, US Department of Health and Human Services (NICHD/NIH/DHHS), Bethesda, Maryland, and Detroit, Michigan, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, Michigan, USA
| | - Derek Miller
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, US Department of Health and Human Services (NICHD/NIH/DHHS), Bethesda, Maryland, and Detroit, Michigan, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, Michigan, USA
| | - Nardhy Gomez-Lopez
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, US Department of Health and Human Services (NICHD/NIH/DHHS), Bethesda, Maryland, and Detroit, Michigan, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, Michigan, USA
- Department of Biochemistry, Microbiology, and Immunology, Wayne State University School of Medicine, Detroit, Michigan, USA
| |
Collapse
|
10
|
Lee JE, Park KH, Kim HJ, Kim YM, Choi JW, Shin S, Lee KN. Proteomic identification of novel plasma biomarkers associated with spontaneous preterm birth in women with preterm labor without infection/inflammation. PLoS One 2021; 16:e0259265. [PMID: 34710180 PMCID: PMC8553083 DOI: 10.1371/journal.pone.0259265] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Accepted: 10/17/2021] [Indexed: 11/18/2022] Open
Abstract
Objective We sought to identify plasma biomarkers associated with spontaneous preterm birth (SPTB, delivery within 21 days of sampling) in women with preterm labor (PTL) without intra-amniotic infection/inflammation (IAI) using label-free quantitative proteomic analysis, as well as to elucidate specific protein pathways involved in these cases. Methods This was a retrospective cohort study comprising 104 singleton pregnant women with PTL (24–32 weeks) who underwent amniocentesis and demonstrated no evidence of IAI. Analysis of pooled plasma samples collected from SPTB cases and term birth (TB) controls (n = 10 for each group) was performed using label-free quantitative mass spectrometry for proteome profiling in a nested case-control study design. Eight candidate proteins of interest were validated by ELISA-based assay and a clot-based assay in the total cohort. Results Ninety-one proteins were differentially expressed (P < 0.05) in plasma samples obtained from SPTB cases, of which 53 (58.2%) were upregulated and 38 (41.8%) were downregulated when compared to TD controls. A validation study confirmed that plasma from women who delivered spontaneously within 21 days of sampling contained significantly higher levels of coagulation factor Ⅴ and lower levels of S100 calcium binding protein A9 (S100A9), especially the former which was independent of baseline variables. The top-ranked pathways related to the 91 differentially expressed proteins were liver-X-receptor/retinoid X receptor (RXR) activation, acute phase response signaling, farnesoid X receptor/RXR activation, coagulation system, and complement system. Conclusions Proteomic analyses in this study identified potential novel biomarkers (i.e., coagulation factor V and S100A9) and potential protein pathways in plasma associated with SPTB in the absence of IAI in women with PTL. The present findings provide novel insights into the molecular pathogenesis and therapeutic targets specific for idiopathic SPTB.
Collapse
Affiliation(s)
- Ji Eun Lee
- Center for Theragnosis, Biomedical Research Division, Korea Institute of Science and Technology, Seoul, Republic of Korea
| | - Kyo Hoon Park
- Department of Obstetrics and Gynecology, Seoul National University College of Medicine, Seoul National University Bundang Hospital, Seongnam, Korea
- * E-mail:
| | - Hyeon Ji Kim
- Department of Obstetrics and Gynecology, Seoul National University College of Medicine, Seoul National University Bundang Hospital, Seongnam, Korea
| | - Yu Mi Kim
- Department of Obstetrics and Gynecology, Seoul National University College of Medicine, Seoul National University Bundang Hospital, Seongnam, Korea
| | - Ji-Woong Choi
- Wide River Institute of Immunology, Seoul National University, Hongcheon, Korea
| | - Sue Shin
- Department of Laboratory Medicine, Seoul National University College of Medicine, Seoul National University Boramae Hospital, Seoul, Korea
| | - Kyong-No Lee
- Department of Obstetrics and Gynecology, Seoul National University College of Medicine, Seoul National University Bundang Hospital, Seongnam, Korea
| |
Collapse
|
11
|
Sabaie H, Dehghani H, Shiva S, Asadi MR, Rezaei O, Taheri M, Rezazadeh M. Mechanistic Insight Into the Regulation of Immune-Related Genes Expression in Autism Spectrum Disorder. Front Mol Biosci 2021; 8:754296. [PMID: 34746237 PMCID: PMC8568055 DOI: 10.3389/fmolb.2021.754296] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Accepted: 10/11/2021] [Indexed: 12/20/2022] Open
Abstract
Autism spectrum disorder (ASD) is a severe neurodevelopmental disorder featuring impairment in verbal and non-verbal interactions, defects in social interactions, stereotypic behaviors as well as restricted interests. In recent times, the incidence of ASD is growing at a rapid pace. In spite of great endeavors devoted to explaining ASD pathophysiology, its precise etiology remains unresolved. ASD pathogenesis is related to different phenomena associated with the immune system; however, the mechanisms behind these immune phenomena as well as the potential contributing genes remain unclear. In the current work, we used a bioinformatics approach to describe the role of long non-coding RNA (lncRNA)-associated competing endogenous RNAs (ceRNAs) in the peripheral blood (PB) samples to figure out the molecular regulatory procedures involved in ASD better. The Gene Expression Omnibus database was used to obtain the PB microarray dataset (GSE89594) from the subjects suffering from ASD and control subjects, containing the data related to both mRNAs and lncRNAs. The list of immune-related genes was obtained from the ImmPort database. In order to determine the immune-related differentially expressed mRNAs (DEmRNAs) and lncRNAs (DElncRNAs), the limma package of R software was used. A protein-protein interaction network was developed for the immune-related DEmRNAs. By employing the Human MicroRNA Disease Database, DIANA-LncBase, and DIANA-TarBase databases, the RNA interaction pairs were determined. We used the Pearson correlation coefficient to discover the positive correlations between DElncRNAs and DEmRNAs within the ceRNA network. Finally, the lncRNA-associated ceRNA network was created based on DElncRNA-miRNA-DEmRNA interactions and co-expression interactions. In addition, the KEGG enrichment analysis was conducted for immune-related DEmRNAs found within the constructed network. This work found four potential DElncRNA-miRNA-DEmRNA axes in ASD pathogenesis, including, LINC00472/hsa-miR-221-3p/PTPN11, ANP32A-IT1/hsa-miR-182-5p/S100A2, LINC00472/hsa-miR-132-3p/S100A2, and RBM26-AS1/hsa-miR-182-5p/S100A2. According to pathway enrichment analysis, the immune-related DEmRNAs were enriched in the "JAK-STAT signaling pathway" and "Adipocytokine signaling pathway." An understanding of regulatory mechanisms of ASD-related immune genes would provide novel insights into the molecular mechanisms behind ASD pathogenesis.
Collapse
Affiliation(s)
- Hani Sabaie
- Molecular Medicine Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Medical Genetics, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Hossein Dehghani
- Department of Molecular Medicine, School of Medicine, Birjand University of Medical Sciences, Birjand, Iran
| | - Shadi Shiva
- Pediatric Health Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mohammad Reza Asadi
- Molecular Medicine Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Medical Genetics, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Omidvar Rezaei
- Skull Base Research Center, Loghman Hakim Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammad Taheri
- Skull Base Research Center, Loghman Hakim Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Institute of Human Genetics, Jena University Hospital, Jena, Germany
| | - Maryam Rezazadeh
- Molecular Medicine Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Medical Genetics, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
12
|
Choltus H, Lavergne M, De Sousa Do Outeiro C, Coste K, Belville C, Blanchon L, Sapin V. Pathophysiological Implication of Pattern Recognition Receptors in Fetal Membranes Rupture: RAGE and NLRP Inflammasome. Biomedicines 2021; 9:biomedicines9091123. [PMID: 34572309 PMCID: PMC8466405 DOI: 10.3390/biomedicines9091123] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Revised: 08/25/2021] [Accepted: 08/26/2021] [Indexed: 12/28/2022] Open
Abstract
Preterm prelabor ruptures of fetal membranes (pPROM) are a pregnancy complication responsible for 30% of all preterm births. This pathology currently appears more as a consequence of early and uncontrolled process runaway activation, which is usually implicated in the physiologic rupture at term: inflammation. This phenomenon can be septic but also sterile. In this latter case, the inflammation depends on some specific molecules called “alarmins” or “damage-associated molecular patterns” (DAMPs) that are recognized by pattern recognition receptors (PRRs), leading to a microbial-free inflammatory response. Recent data clarify how this activation works and which receptor translates this inflammatory signaling into fetal membranes (FM) to manage a successful rupture after 37 weeks of gestation. In this context, this review focused on two PRRs: the receptor for advanced glycation end-products (RAGE) and the NLRP7 inflammasome.
Collapse
Affiliation(s)
- Helena Choltus
- CNRS, INSERM, GReD, Université Clermont Auvergne, 63000 Clermont-Ferrand, France; (H.C.); (M.L.); (C.D.S.D.O.); (K.C.); (C.B.); (L.B.)
| | - Marilyne Lavergne
- CNRS, INSERM, GReD, Université Clermont Auvergne, 63000 Clermont-Ferrand, France; (H.C.); (M.L.); (C.D.S.D.O.); (K.C.); (C.B.); (L.B.)
| | - Coraline De Sousa Do Outeiro
- CNRS, INSERM, GReD, Université Clermont Auvergne, 63000 Clermont-Ferrand, France; (H.C.); (M.L.); (C.D.S.D.O.); (K.C.); (C.B.); (L.B.)
| | - Karen Coste
- CNRS, INSERM, GReD, Université Clermont Auvergne, 63000 Clermont-Ferrand, France; (H.C.); (M.L.); (C.D.S.D.O.); (K.C.); (C.B.); (L.B.)
| | - Corinne Belville
- CNRS, INSERM, GReD, Université Clermont Auvergne, 63000 Clermont-Ferrand, France; (H.C.); (M.L.); (C.D.S.D.O.); (K.C.); (C.B.); (L.B.)
| | - Loïc Blanchon
- CNRS, INSERM, GReD, Université Clermont Auvergne, 63000 Clermont-Ferrand, France; (H.C.); (M.L.); (C.D.S.D.O.); (K.C.); (C.B.); (L.B.)
| | - Vincent Sapin
- CNRS, INSERM, GReD, Université Clermont Auvergne, 63000 Clermont-Ferrand, France; (H.C.); (M.L.); (C.D.S.D.O.); (K.C.); (C.B.); (L.B.)
- CHU de Clermont-Ferrand, Biochemistry and Molecular Genetic Department, 63000 Clermont-Ferrand, France
- Correspondence: ; Tel.: +33-473-178-174
| |
Collapse
|
13
|
Zhang Y, Yang X, Zhu XL, Bai H, Wang ZZ, Zhang JJ, Hao CY, Duan HB. S100A gene family: immune-related prognostic biomarkers and therapeutic targets for low-grade glioma. Aging (Albany NY) 2021; 13:15459-15478. [PMID: 34148033 PMCID: PMC8221329 DOI: 10.18632/aging.203103] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Accepted: 05/13/2021] [Indexed: 02/06/2023]
Abstract
BACKGROUND Despite the better prognosis given by surgical resection and chemotherapy in low-grade glioma (LGG), progressive transformation is still a huge concern. In this case, the S100A gene family, being capable of regulating inflammatory responses, can promote tumor development. METHODS The analysis was carried out via ONCOMINE, GEPIA, cBioPortal, String, GeneMANIA, WebGestalt, LinkedOmics, TIMER, CGGA, R 4.0.2 and immunohistochemistry. RESULTS S100A2, S100A6, S100A10, S100A11, and S100A16 were up-regulated and S100A1 and S100A13 were down-regulated in LGG compared to normal tissues. S100A3, S100A4, S100A8, and S100A9 expression was up-regulated during the progression of glioma grade. In addition, genetic variation of the S100A family was high in LGG, and the S100A family genes mostly function through IL-17 signaling pathway, S100 binding protein, and inflammatory responses. The TIMER database also revealed a relationship between gene expression and immune cell infiltration. High expression of S100A2, S100A3, S100A4, S100A6, S100A8, S100A9, S100A10, S100A11, S100A13, and S100A16 was significantly associated with poor prognosis in LGG patients. S100A family genes S100A2, S100A3, S100A6, S100A10, and S100A11 may be prognosis-related genes in LGG, and were significantly associated with IDH mutation and 1p19q codeletion. The immunohistochemical staining results also confirmed that S100A2, S100A3, S100A6, S100A10, and S100A11 expression was upregulated in LGG. CONCLUSION The S100A family plays a vital role in LGG pathogenesis, presumably facilitating LGG progression via modulating inflammatory state and immune cell infiltration.
Collapse
Affiliation(s)
- Yu Zhang
- Department of Neurosurgery, First Hospital of Shanxi Medical University, Taiyuan 030001, Shanxi, P.R. China
| | - Xin Yang
- Department of Neurosurgery, First Hospital of Shanxi Medical University, Taiyuan 030001, Shanxi, P.R. China
| | - Xiao-Lin Zhu
- Department of Neurosurgery, First Hospital of Shanxi Medical University, Taiyuan 030001, Shanxi, P.R. China
| | - Hao Bai
- Department of Neurosurgery, First Hospital of Shanxi Medical University, Taiyuan 030001, Shanxi, P.R. China
| | - Zhuang-Zhuang Wang
- Department of Neurosurgery, First Hospital of Shanxi Medical University, Taiyuan 030001, Shanxi, P.R. China
| | - Jun-Jie Zhang
- Department of Neurosurgery, First Hospital of Shanxi Medical University, Taiyuan 030001, Shanxi, P.R. China
| | - Chun-Yan Hao
- Department of Geriatrics, First Hospital of Shanxi Medical University, Taiyuan 030001, Shanxi, P.R. China
| | - Hu-Bin Duan
- Department of Neurosurgery, First Hospital of Shanxi Medical University, Taiyuan 030001, Shanxi, P.R. China.,Department of Neurosurgery, Lvliang People's Hospital, Lvliang 033000, Shanxi, P.R. China
| |
Collapse
|