1
|
Svecla M, Li-Gao R, Falck D, Bonacina F. N-glycosylation signature and its relevance in cardiovascular immunometabolism. Vascul Pharmacol 2025; 159:107474. [PMID: 39988310 DOI: 10.1016/j.vph.2025.107474] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Revised: 01/21/2025] [Accepted: 02/20/2025] [Indexed: 02/25/2025]
Abstract
Glycosylation is a post-translational modification in which complex, branched carbohydrates (glycans) are covalently attached to proteins or lipids. Asparagine-link protein (N-) glycosylation is among the most common types of glycosylation. This process is essential for many biological and cellular functions, and impaired N-glycosylation has been widely implicated in inflammation and cardiovascular diseases. Different technical approaches have been used to increase the coverage of the N-glycome, revealing a high level of complexity of glycans, regarding their structure and attachment site on a protein. In this context, new insights from genomic studies have revealed a genetic regulation of glycosylation, linking genetic variants to total plasma N-glycosylation and N-glycosylation of immunoglobulin G (IgG). In addition, RNAseq approaches have revealed a degree of transcriptional regulation for the glycoenzymes involved in glycan structure. However, our understanding of the association between cardiovascular risk and glycosylation, determined by a complex overlay of genetic and environmental factors, remains limited. Mostly, plasma N-glycosylation profiling in different human cohorts or experimental investigations of specific enzyme functions in models of atherosclerosis have been reported. Most of the uncovered glycosylation associations with pathological mechanisms revolve around the recruitment of inflammatory cells to the vessel wall and lipoprotein metabolism. This review aims to summarise insights from omics studies into the immune and metabolic regulation of N-glycosylation and its association with cardiovascular and metabolic disease risk and to provide mechanistic insights from experimental models. The combination of emerging techniques for glycomics and glycoproteomics with already achieved omics approaches to map the transcriptomic, epigenomic, and metabolomic profile at single-cell resolution will deepen our understanding of the molecular regulation of glycosylation as well as identify novel biomarkers and targets for cardiovascular disease prevention and treatment.
Collapse
Affiliation(s)
- Monika Svecla
- Department of Neurosurgery, Charité Universitätsmedizin Berlin, Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Ruifang Li-Gao
- Department of Clinical Epidemiology, Leiden University Medical Center, Leiden, the Netherlands
| | - David Falck
- Leiden University Medical Center, Center for Proteomics and Metabolomics, Glycomics Group, Leiden, the Netherlands
| | - Fabrizia Bonacina
- Department of Excellence of Pharmacological and Biomolecular Sciences "Rodolfo Paoletti", University of Milan, Milan, Italy.
| |
Collapse
|
2
|
Qian R, Zhao X, Lyu D, Xu Q, Yuan K, Luo X, Wang W, Wang Y, Liu Y, Cheng Y, Tan Y, Mou F, Yuan C, Yu S. Identification of Causal Genes and Potential Drug Targets for Restless Legs Syndrome: A Comprehensive Mendelian Randomization Study. Pharmaceuticals (Basel) 2024; 17:1626. [PMID: 39770468 PMCID: PMC11728827 DOI: 10.3390/ph17121626] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2024] [Revised: 11/22/2024] [Accepted: 11/28/2024] [Indexed: 01/16/2025] Open
Abstract
Background: Restless legs syndrome (RLS) is a common sensorimotor sleep disorder that affects sleep quality of life. Much effort has been made to make progress in RLS pharmacotherapy; however, patients with RLS still report poor long-term symptom control. Methods: Comprehensive Mendelian randomization (MR) was performed to search for potential causal genes and drug targets using the cis-pQTL and RLS GWAS data. Robustness was validated using the summary-based Mendelian randomization (SMR) method and co-localization analysis. Further evidence of pleiotropy of the target genes and their potential side effects was provided by phenome-wide MR analysis (MR-PheWAS). Finally, molecular docking simulations were conducted on drug candidates corresponding to these targets, which revealed promising binding affinities and interaction patterns and underscored the druggable potential of the target gene. All of the analyses above were conducted in the context of Homo sapiens. Results:MAN1A2 showed a statistically significant result in the MR analysis, which was validated through SMR and co-localization analysis. The MR-PheWAS showed a low probability of pleiotropy and prospective side effects. Molecular docking was used to visualize the binding structure and fine affinity for MAN1A2 and the drugs predicted by DSigDB. Conclusions: Our study provides comprehensive evidence supporting MAN1A2 as a promising causal gene and therapeutic target for RLS, offering insights into the underlying molecular mechanisms and paving the way for future drug development efforts.
Collapse
Affiliation(s)
- Ruiyi Qian
- Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai 200030, China; (R.Q.); (X.Z.); (D.L.); (Q.X.); (X.L.); (W.W.); (Y.W.); (Y.L.); (Y.C.); (Y.T.); (F.M.)
| | - Xue Zhao
- Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai 200030, China; (R.Q.); (X.Z.); (D.L.); (Q.X.); (X.L.); (W.W.); (Y.W.); (Y.L.); (Y.C.); (Y.T.); (F.M.)
| | - Dongbin Lyu
- Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai 200030, China; (R.Q.); (X.Z.); (D.L.); (Q.X.); (X.L.); (W.W.); (Y.W.); (Y.L.); (Y.C.); (Y.T.); (F.M.)
| | - Qingqing Xu
- Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai 200030, China; (R.Q.); (X.Z.); (D.L.); (Q.X.); (X.L.); (W.W.); (Y.W.); (Y.L.); (Y.C.); (Y.T.); (F.M.)
| | - Kai Yuan
- State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, Institute of Digestive Disease, Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong SAR 999077, China;
| | - Xin Luo
- Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai 200030, China; (R.Q.); (X.Z.); (D.L.); (Q.X.); (X.L.); (W.W.); (Y.W.); (Y.L.); (Y.C.); (Y.T.); (F.M.)
| | - Wanying Wang
- Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai 200030, China; (R.Q.); (X.Z.); (D.L.); (Q.X.); (X.L.); (W.W.); (Y.W.); (Y.L.); (Y.C.); (Y.T.); (F.M.)
| | - Yang Wang
- Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai 200030, China; (R.Q.); (X.Z.); (D.L.); (Q.X.); (X.L.); (W.W.); (Y.W.); (Y.L.); (Y.C.); (Y.T.); (F.M.)
| | - Yutong Liu
- Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai 200030, China; (R.Q.); (X.Z.); (D.L.); (Q.X.); (X.L.); (W.W.); (Y.W.); (Y.L.); (Y.C.); (Y.T.); (F.M.)
| | - Yu Cheng
- Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai 200030, China; (R.Q.); (X.Z.); (D.L.); (Q.X.); (X.L.); (W.W.); (Y.W.); (Y.L.); (Y.C.); (Y.T.); (F.M.)
| | - Yingting Tan
- Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai 200030, China; (R.Q.); (X.Z.); (D.L.); (Q.X.); (X.L.); (W.W.); (Y.W.); (Y.L.); (Y.C.); (Y.T.); (F.M.)
| | - Fan Mou
- Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai 200030, China; (R.Q.); (X.Z.); (D.L.); (Q.X.); (X.L.); (W.W.); (Y.W.); (Y.L.); (Y.C.); (Y.T.); (F.M.)
| | - Chengmei Yuan
- Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai 200030, China; (R.Q.); (X.Z.); (D.L.); (Q.X.); (X.L.); (W.W.); (Y.W.); (Y.L.); (Y.C.); (Y.T.); (F.M.)
| | - Shunying Yu
- Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai 200030, China; (R.Q.); (X.Z.); (D.L.); (Q.X.); (X.L.); (W.W.); (Y.W.); (Y.L.); (Y.C.); (Y.T.); (F.M.)
| |
Collapse
|
3
|
Batara DC, Kim HJ, Phan LT, Kim M, Son YO, Lee S, Park SI, Choi YS, Beck S, Kim SH. Elevated α-1,2-mannosidase MAN1C1 in glioma stem cells and its implications for immunological changes and prognosis in glioma patients. Sci Rep 2024; 14:22159. [PMID: 39333557 PMCID: PMC11436702 DOI: 10.1038/s41598-024-72901-2] [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: 05/31/2024] [Accepted: 09/11/2024] [Indexed: 09/29/2024] Open
Abstract
Glioblastoma multiforme (GBM) is the most aggressive type of primary brain tumor, and the presence of glioma stem cells (GSCs) has been linked to its resistance to treatments and recurrence. Additionally, aberrant glycosylation has been implicated in the aggressiveness of cancers. However, the influence and underlying mechanism of N-glycosylation on the GSC phenotype and GBM malignancy remain elusive. Here, we performed an in-silico analysis approach on publicly available datasets to examine the function of N-glycosylation-related genes in GSCs and gliomas, accompanied by a qRT-PCR validation experiment. We found that high α-1,2-mannosidase MAN1C1 is associated with immunological functions and worse survival of glioma patients. Differential gene expression analysis and qRT-PCR validation revealed that MAN1C1 is highly expressed in GSCs. Furthermore, higher MAN1C1 expression predicts worse outcomes in glioma patients. Also, MAN1C1 expression is increased in the perinecrotic region of GBM and is associated with immunological and inflammatory functions, a hallmark of the GBM mesenchymal subtype. Further analysis confirmed that MAN1C1 expression is closely associated with infiltrating immune cells and disrupted immune response in the GBM microenvironment. These suggest that MAN1C1 is a potential biomarker for gliomas and may be important as an immunotherapeutic target for GBM.
Collapse
Affiliation(s)
- Don Carlo Batara
- Animal Molecular Biochemistry Laboratory, Department of Animal Science, College of Agriculture and Life Sciences, Chonnam National University, Gwangju, 61186, Republic of Korea
| | - Hyun-Jin Kim
- Animal Molecular Biochemistry Laboratory, Department of Animal Science, College of Agriculture and Life Sciences, Chonnam National University, Gwangju, 61186, Republic of Korea
| | - Le Thi Phan
- Animal Molecular Biochemistry Laboratory, Department of Animal Science, College of Agriculture and Life Sciences, Chonnam National University, Gwangju, 61186, Republic of Korea
- Computational Biology and Bioinformatics Laboratory, Department of Integrative Biotechnology, Sungkyunkwan University, Suwon, Gyeonggi-do, 16419, Republic of Korea
| | - Minseo Kim
- Animal Molecular Biochemistry Laboratory, Department of Animal Science, College of Agriculture and Life Sciences, Chonnam National University, Gwangju, 61186, Republic of Korea
| | - Young-Ok Son
- Department of Animal Biotechnology, Faculty of Biotechnology, College of Applied Life Sciences, Interdisciplinary Graduate Program in Advanced Convergence Technology and Science, Jeju National University, Jeju, 63243, Republic of Korea
| | - Seongsoo Lee
- Gwangju Center, Korea Basic Science Institute (KBSI), 49, Dosicheomdansaneop-ro, Nam-gu, Gwangju, 61751, Republic of Korea
- Department of Systems Biotechnology, Chung-Ang University, Anseong-si, Gyeonggi-do, 17546, Republic of Korea
| | - Sang-Ik Park
- Laboratory of Veterinary Pathology, College of Veterinary Medicine and BK21 Plus Project Team, Chonnam National University, Gwangju, 61186, Republic of Korea
| | - Young Sun Choi
- Jeollanam-do Agriculture Research and Extension Services Livestock Research Institute, Naju-si, Jeollanam-do, 58213, Republic of Korea
| | - Samuel Beck
- Department of Dermatology, Center for Aging Research, Chobanian & Avedisian School of Medicine, Boston University, Boston, 02118, USA.
| | - Sung-Hak Kim
- Animal Molecular Biochemistry Laboratory, Department of Animal Science, College of Agriculture and Life Sciences, Chonnam National University, Gwangju, 61186, Republic of Korea.
| |
Collapse
|
4
|
Chatham JC, Patel RP. Protein glycosylation in cardiovascular health and disease. Nat Rev Cardiol 2024; 21:525-544. [PMID: 38499867 DOI: 10.1038/s41569-024-00998-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 02/13/2024] [Indexed: 03/20/2024]
Abstract
Protein glycosylation, which involves the attachment of carbohydrates to proteins, is one of the most abundant protein co-translational and post-translational modifications. Advances in technology have substantially increased our knowledge of the biosynthetic pathways involved in protein glycosylation, as well as how changes in glycosylation can affect cell function. In addition, our understanding of the role of protein glycosylation in disease processes is growing, particularly in the context of immune system function, infectious diseases, neurodegeneration and cancer. Several decades ago, cell surface glycoproteins were found to have an important role in regulating ion transport across the cardiac sarcolemma. However, with very few exceptions, our understanding of how changes in protein glycosylation influence cardiovascular (patho)physiology remains remarkably limited. Therefore, in this Review, we aim to provide an overview of N-linked and O-linked protein glycosylation, including intracellular O-linked N-acetylglucosamine protein modification. We discuss our current understanding of how all forms of protein glycosylation contribute to normal cardiovascular function and their roles in cardiovascular disease. Finally, we highlight potential gaps in our knowledge about the effects of protein glycosylation on the heart and vascular system, highlighting areas for future research.
Collapse
Affiliation(s)
- John C Chatham
- Division of Molecular and Cellular Pathology, Department of Pathology, University of Alabama at Birmingham, Birmingham, AL, USA.
| | - Rakesh P Patel
- Division of Molecular and Cellular Pathology, Department of Pathology, University of Alabama at Birmingham, Birmingham, AL, USA
| |
Collapse
|
5
|
Alfaro GF, Palombo V, D’Andrea M, Cao W, Zhang Y, Beever JE, Muntifering RB, Pacheco WJ, Rodning SP, Wang X, Moisá SJ. Hepatic transcript profiling in beef cattle: Effects of feeding endophyte-infected tall fescue seeds. PLoS One 2024; 19:e0306431. [PMID: 39058685 PMCID: PMC11280227 DOI: 10.1371/journal.pone.0306431] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2024] [Accepted: 06/17/2024] [Indexed: 07/28/2024] Open
Abstract
The objective of our study was to evaluate the effect of endophyte-infected tall fescue (E+) seeds intake on liver tissue transcriptome in growing Angus × Simmental steers and heifers through RNA-seq analysis. Normal weaned calves (~8 months old) received either endophyte-free tall fescue (E-; n = 3) or infected tall fescue (E+; n = 6) seeds for a 30-d period. The diet offered was ad libitum bermudagrass (Cynodon dactylon) hay combined with a nutritional supplement of 1.61 kg (DM basis) of E+ or E- tall fescue seeds, and 1.61 kg (DM basis) of energy/protein supplement pellets for a 30-d period. Dietary E+ tall fescue seeds were included in a rate of 20 μg of ergovaline/kg BW/day. Liver tissue was individually obtained through biopsy at d 30. After preparation and processing of the liver samples for RNA sequencing, we detected that several metabolic pathways were activated (i.e., upregulated) by the consumption of E+ tall fescue. Among them, oxidative phosphorylation, ribosome biogenesis, protein processing in endoplasmic reticulum and apoptosis, suggesting an active mechanism to cope against impairment in normal liver function. Interestingly, hepatic protein synthesis might increase due to E+ consumption. In addition, there was upregulation of "thermogenesis" KEGG pathway, showing a possible increase in energy expenditure in liver tissue due to consumption of E+ diet. Therefore, results from our study expand the current knowledge related to liver metabolism of growing beef cattle under tall fescue toxicosis.
Collapse
Affiliation(s)
- Gastón F. Alfaro
- Department of Animal Sciences, Auburn University, Auburn, AL, United States of America
| | - Valentino Palombo
- Department of Agricultural, Environmental and Food Sciences, Università degli Studi del Molise, Campobasso, Italy
| | - MariaSilvia D’Andrea
- Department of Agricultural, Environmental and Food Sciences, Università degli Studi del Molise, Campobasso, Italy
| | - Wenqi Cao
- Department of Pathobiology, College of Veterinary Medicine, Auburn University, Auburn, AL, United States of America
| | - Yue Zhang
- Department of Pathobiology, College of Veterinary Medicine, Auburn University, Auburn, AL, United States of America
| | - Jonathan E. Beever
- Department of Animal Sciences, University of Tennessee, Knoxville, TN, United States of America
| | | | - Wilmer J. Pacheco
- Department of Poultry Sciences, Auburn University, Auburn, AL, United States of America
| | - Soren P. Rodning
- Department of Animal Sciences, Auburn University, Auburn, AL, United States of America
| | - Xu Wang
- Department of Pathobiology, College of Veterinary Medicine, Auburn University, Auburn, AL, United States of America
- HudsonAlpha Institute for Biotechnology, Huntsville, AL, United States of America
| | - Sonia J. Moisá
- Department of Animal Sciences, University of Tennessee, Knoxville, TN, United States of America
| |
Collapse
|
6
|
Yu Y, Hua J, Chen L. Autophagy-related molecular clusters identified as indicators for distinguishing active and latent TB infection in pediatric patients. BMC Pediatr 2024; 24:398. [PMID: 38890657 PMCID: PMC11186109 DOI: 10.1186/s12887-024-04881-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/27/2024] [Accepted: 06/11/2024] [Indexed: 06/20/2024] Open
Abstract
BACKGROUND Autophagy is crucial for controlling the manifestation of tuberculosis. This study intends to discover autophagy-related molecular clusters as biomarkers for discriminating between latent tuberculosis (LTBI) and active tuberculosis (ATB) in children through gene expression profile analysis. METHODS The expression of autophagy modulators was examined in pediatric patients with LTBI and ATB utilizing public datasets from the Gene Expression Omnibus (GEO) collection (GSE39939 and GSE39940). RESULTS In a training dataset (GSE39939), patients with LTBI and ATB exhibited the expression of autophagy-related genes connected with their active immune responses. Two molecular clusters associated with autophagy were identified. Compared to Cluster 1, Cluster 2 was distinguished through decreased adaptive cellular immune response and enhanced inflammatory activation, according to single-sample gene set enrichment analysis (ssGSEA). Per the study of gene set variation, Cluster 2's differentially expressed genes (DEGs) played a role in synthesizing transfer RNA, DNA repair and recombination, and primary immunodeficiency. The peak variation efficiency, root mean square error, and area under the curve (AUC) (AUC = 0.950) were all lowered in random forest models. Finally, a seven-gene-dependent random forest profile was created utilizing the CD247, MAN1C1, FAM84B, HSZFP36, SLC16A10, DTX3, and SIRT4 genes, which performed well against the validation dataset GSE139940 (AUC = 0.888). The nomogram calibration and decision curves performed well in identifying ATB from LTBI. CONCLUSIONS In summary, according to the present investigation, autophagy and the immunopathology of TB might be correlated. Furthermore, this investigation established a compelling prediction expression profile for measuring autophagy subtype development risks, which might be employed as possible biomarkers in children to differentiate ATB from LTBI.
Collapse
Affiliation(s)
- Yang Yu
- Department of Pediatric, Nanjing Lishui People's Hospital, Zhongda Hospital Lishui Branch, Southeast University, Nanjing, China
| | - Jie Hua
- Department of Gastroenterology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Liang Chen
- Department of Infectious Diseases, Taikang Xianlin Drum Tower Hospital, Affiliated Hospital of Medical College of Nanjing University, Qixia District, NO 188, Lingshan North Road, Qixia District, Nanjing, 210046, China.
| |
Collapse
|
7
|
Xu X, Yin K, Wu R. Systematic Investigation of the Trafficking of Glycoproteins on the Cell Surface. Mol Cell Proteomics 2024; 23:100761. [PMID: 38593903 PMCID: PMC11087972 DOI: 10.1016/j.mcpro.2024.100761] [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: 02/22/2024] [Revised: 03/30/2024] [Accepted: 04/03/2024] [Indexed: 04/11/2024] Open
Abstract
Glycoproteins located on the cell surface play a pivotal role in nearly every extracellular activity. N-glycosylation is one of the most common and important protein modifications in eukaryotic cells, and it often regulates protein folding and trafficking. Glycosylation of cell-surface proteins undergoes meticulous regulation by various enzymes in the endoplasmic reticulum (ER) and the Golgi, ensuring their proper folding and trafficking to the cell surface. However, the impacts of protein N-glycosylation, N-glycan maturity, and protein folding status on the trafficking of cell-surface glycoproteins remain to be explored. In this work, we comprehensively and site-specifically studied the trafficking of cell-surface glycoproteins in human cells. Integrating metabolic labeling, bioorthogonal chemistry, and multiplexed proteomics, we investigated 706 N-glycosylation sites on 396 cell-surface glycoproteins in monocytes, either by inhibiting protein N-glycosylation, disturbing N-glycan maturation, or perturbing protein folding in the ER. The current results reveal their distinct impacts on the trafficking of surface glycoproteins. The inhibition of protein N-glycosylation dramatically suppresses the trafficking of many cell-surface glycoproteins. The N-glycan immaturity has more substantial effects on proteins with high N-glycosylation site densities, while the perturbation of protein folding in the ER exerts a more pronounced impact on surface glycoproteins with larger sizes. Furthermore, for N-glycosylated proteins, their trafficking to the cell surface is related to the secondary structures and adjacent amino acid residues of glycosylation sites. Systematic analysis of surface glycoprotein trafficking advances our understanding of the mechanisms underlying protein secretion and surface presentation.
Collapse
Affiliation(s)
- Xing Xu
- School of Chemistry and Biochemistry and the Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, Georgia, USA
| | - Kejun Yin
- School of Chemistry and Biochemistry and the Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, Georgia, USA
| | - Ronghu Wu
- School of Chemistry and Biochemistry and the Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, Georgia, USA.
| |
Collapse
|
8
|
Teng D, Wang W, Jia W, Song J, Gong L, Zhong L, Yang J. The effects of glycosylation modifications on monocyte recruitment and foam cell formation in atherosclerosis. Biochim Biophys Acta Mol Basis Dis 2024; 1870:167027. [PMID: 38237743 DOI: 10.1016/j.bbadis.2024.167027] [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: 09/16/2023] [Revised: 01/10/2024] [Accepted: 01/10/2024] [Indexed: 01/23/2024]
Abstract
The monocyte recruitment and foam cell formation have been intensively investigated in atherosclerosis. Nevertheless, as the study progressed, it was obvious that crucial molecules participated in the monocyte recruitment and the membrane proteins in macrophages exhibited substantial glycosylation modifications. These modifications can exert a significant influence on protein functions and may even impact the overall progression of diseases. This article provides a review of the effects of glycosylation modifications on monocyte recruitment and foam cell formation. By elaborating on these effects, we aim to understand the underlying mechanisms of atherogenesis further and to provide new insights into the future treatment of atherosclerosis.
Collapse
Affiliation(s)
- Da Teng
- Yantai Yuhuangding Hospital affiliated to Qingdao University, Yantai, Shandong, People's Republic of China; Qingdao University, Qingdao, Shandong, People's Republic of China
| | - Wenlong Wang
- Yantai Yuhuangding Hospital affiliated to Qingdao University, Yantai, Shandong, People's Republic of China; Qingdao University, Qingdao, Shandong, People's Republic of China
| | - Wenjuan Jia
- Yantai Yuhuangding Hospital affiliated to Qingdao University, Yantai, Shandong, People's Republic of China; Qingdao University, Qingdao, Shandong, People's Republic of China
| | - Jikai Song
- Yantai Yuhuangding Hospital affiliated to Qingdao University, Yantai, Shandong, People's Republic of China; Qingdao University, Qingdao, Shandong, People's Republic of China
| | - Lei Gong
- Yantai Yuhuangding Hospital affiliated to Qingdao University, Yantai, Shandong, People's Republic of China
| | - Lin Zhong
- Yantai Yuhuangding Hospital affiliated to Qingdao University, Yantai, Shandong, People's Republic of China.
| | - Jun Yang
- Yantai Yuhuangding Hospital affiliated to Qingdao University, Yantai, Shandong, People's Republic of China; Qingdao University, Qingdao, Shandong, People's Republic of China.
| |
Collapse
|
9
|
Luan J, Ji X, Liu L. PPARγ in Atherosclerotic Endothelial Dysfunction: Regulatory Compounds and PTMs. Int J Mol Sci 2023; 24:14494. [PMID: 37833942 PMCID: PMC10572723 DOI: 10.3390/ijms241914494] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Revised: 09/20/2023] [Accepted: 09/22/2023] [Indexed: 10/15/2023] Open
Abstract
The formation of atherosclerotic plaques is one of the main sources of cardiovascular disease. In addition to known risk factors such as dyslipidemia, diabetes, obesity, and hypertension, endothelial dysfunction has been shown to play a key role in the formation and progression of atherosclerosis. Peroxisome proliferator-activated receptor-gamma (PPARγ), a transcription factor belonging to the steroid superfamily, is expressed in the aorta and plays a critical role in protecting endothelial function. It thereby serves as a target for treating both diabetes and atherosclerosis. Although many studies have examined endothelial cell disorders in atherosclerosis, the role of PPARγ in endothelial dysfunction is still not well understood. In this review, we summarize the possible mechanisms of action behind PPARγ regulatory compounds and post-translational modifications (PTMs) of PPARγ in the control of endothelial function. We also explore the potential use of endothelial PPARγ-targeted agents in the prevention and treatment of atherosclerosis.
Collapse
Affiliation(s)
| | | | - Longhua Liu
- School of Exercise and Health, Shanghai University of Sport, Shanghai 200082, China
| |
Collapse
|
10
|
Freise C, Zappe A, Löwa N, Schnorr J, Pagel K, Wiekhorst F, Taupitz M. Uremic Toxin-Induced Exosome-like Extracellular Vesicles Contain Enhanced Levels of Sulfated Glycosaminoglycans which Facilitate the Interaction with Very Small Superparamagnetic Iron Oxide Particles. Int J Mol Sci 2023; 24:14253. [PMID: 37762555 PMCID: PMC10532171 DOI: 10.3390/ijms241814253] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Revised: 09/11/2023] [Accepted: 09/15/2023] [Indexed: 09/29/2023] Open
Abstract
Uremic toxins exert pathophysiological effects on cells and tissues, such as the generation of a pro-calcifying subtype of exosome-like extracellular vesicles (EVs) in vascular cells. Little is known about the effects of the toxins on the surface structure of EVs. Thus, we studied the effects of uremic toxins on the abundance of sulfated glycosaminoglycans (GAGs) in EVs, and the implications for binding of ligands such as very small superparamagnetic iron oxide particles (VSOPs) which could be of relevance for radiological EV-imaging. Vascular cells were treated with the uremic toxins NaH2PO4 and a mixture of urea and indoxyl sulfate. Uremia in rats was induced by adenine feeding. EVs were isolated from culture supernatants and plasma of rats. By proton T1-relaxometry, magnetic particle spectroscopy, and analysis of genes, proteins, and GAG-contents, we analyzed the roles of GAGs in the ligand binding of EVs. By influencing GAG-associated genes in host cells, uremic toxins induced higher GAG contents in EVs, particularly of sulfated chondroitin sulfate and heparan sulfate chains. EVs with high GAG content interacted stronger with VSOPs compared to control ones. This was confirmed by experiments with GAG-depleted EVs from genetically modified CHO cells and with uremic rat-derived EVs. Mechanistically, uremic toxin-induced PI3K/AKT-signaling and expression of the sulfate transporter SLC26A2 in host cells contributed to high GAG contents in EVs. In conclusion, uremic conditions induce enhanced GAG contents in EVs, which entails a stronger interaction with VSOPs. VSOPs might be suitable for radiological imaging of EVs rich in GAGs.
Collapse
Affiliation(s)
- Christian Freise
- Department of Radiology, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Charitéplatz 1, 10117 Berlin, Germany; (J.S.); (M.T.)
| | - Andreas Zappe
- Department of Biology, Chemistry and Pharmacy, Freie Universität Berlin, Altensteinstraße 23A, 14195 Berlin, Germany; (A.Z.); (K.P.)
| | - Norbert Löwa
- Metrology for Magnetic Nanoparticles Berlin, Physikalisch-Technische Bundesanstalt Berlin, Abbestr. 2, 10587 Berlin, Germany; (N.L.); (F.W.)
| | - Jörg Schnorr
- Department of Radiology, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Charitéplatz 1, 10117 Berlin, Germany; (J.S.); (M.T.)
| | - Kevin Pagel
- Department of Biology, Chemistry and Pharmacy, Freie Universität Berlin, Altensteinstraße 23A, 14195 Berlin, Germany; (A.Z.); (K.P.)
| | - Frank Wiekhorst
- Metrology for Magnetic Nanoparticles Berlin, Physikalisch-Technische Bundesanstalt Berlin, Abbestr. 2, 10587 Berlin, Germany; (N.L.); (F.W.)
| | - Matthias Taupitz
- Department of Radiology, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Charitéplatz 1, 10117 Berlin, Germany; (J.S.); (M.T.)
| |
Collapse
|
11
|
Erger F, Aryal RP, Reusch B, Matsumoto Y, Meyer R, Zeng J, Knopp C, Noel M, Muerner L, Wenzel A, Kohl S, Tschernoster N, Rappl G, Rouvet I, Schröder-Braunstein J, Seibert FS, Thiele H, Häusler MG, Weber LT, Büttner-Herold M, Elbracht M, Cummings SF, Altmüller J, Habbig S, Cummings RD, Beck BB. Germline C1GALT1C1 mutation causes a multisystem chaperonopathy. Proc Natl Acad Sci U S A 2023; 120:e2211087120. [PMID: 37216524 PMCID: PMC10235935 DOI: 10.1073/pnas.2211087120] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Accepted: 03/28/2023] [Indexed: 05/24/2023] Open
Abstract
Mutations in genes encoding molecular chaperones can lead to chaperonopathies, but none have so far been identified causing congenital disorders of glycosylation. Here we identified two maternal half-brothers with a novel chaperonopathy, causing impaired protein O-glycosylation. The patients have a decreased activity of T-synthase (C1GALT1), an enzyme that exclusively synthesizes the T-antigen, a ubiquitous O-glycan core structure and precursor for all extended O-glycans. The T-synthase function is dependent on its specific molecular chaperone Cosmc, which is encoded by X-chromosomal C1GALT1C1. Both patients carry the hemizygous variant c.59C>A (p.Ala20Asp; A20D-Cosmc) in C1GALT1C1. They exhibit developmental delay, immunodeficiency, short stature, thrombocytopenia, and acute kidney injury (AKI) resembling atypical hemolytic uremic syndrome. Their heterozygous mother and maternal grandmother show an attenuated phenotype with skewed X-inactivation in blood. AKI in the male patients proved fully responsive to treatment with the complement inhibitor Eculizumab. This germline variant occurs within the transmembrane domain of Cosmc, resulting in dramatically reduced expression of the Cosmc protein. Although A20D-Cosmc is functional, its decreased expression, though in a cell or tissue-specific manner, causes a large reduction of T-synthase protein and activity, which accordingly leads to expression of varied amounts of pathological Tn-antigen (GalNAcα1-O-Ser/Thr/Tyr) on multiple glycoproteins. Transient transfection of patient lymphoblastoid cells with wild-type C1GALT1C1 partially rescued the T-synthase and glycosylation defect. Interestingly, all four affected individuals have high levels of galactose-deficient IgA1 in sera. These results demonstrate that the A20D-Cosmc mutation defines a novel O-glycan chaperonopathy and causes the altered O-glycosylation status in these patients.
Collapse
Affiliation(s)
- Florian Erger
- Institute of Human Genetics, University Hospital Cologne, Faculty of Medicine, University of Cologne, 50931Cologne, Germany
- Center for Molecular Medicine Cologne, University of Cologne, 50931Cologne, Germany
| | - Rajindra P. Aryal
- Division of Surgical Sciences, Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA02215
| | - Björn Reusch
- Institute of Human Genetics, University Hospital Cologne, Faculty of Medicine, University of Cologne, 50931Cologne, Germany
- Center for Molecular Medicine Cologne, University of Cologne, 50931Cologne, Germany
| | - Yasuyuki Matsumoto
- Division of Surgical Sciences, Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA02215
| | - Robert Meyer
- Institute for Human Genetics and Genomic Medicine, Medical Faculty, Rheinisch-Westfälische Technische Hochschule Aachen University, 52074Aachen, Germany
| | - Junwei Zeng
- Division of Surgical Sciences, Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA02215
- Medical Research Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, 510080Guangzhou, China
| | - Cordula Knopp
- Institute for Human Genetics and Genomic Medicine, Medical Faculty, Rheinisch-Westfälische Technische Hochschule Aachen University, 52074Aachen, Germany
| | - Maxence Noel
- Division of Surgical Sciences, Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA02215
| | - Lukas Muerner
- Division of Surgical Sciences, Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA02215
- Institute of Pharmacology, University of Bern, 3010Bern, Switzerland
| | - Andrea Wenzel
- Institute of Human Genetics, University Hospital Cologne, Faculty of Medicine, University of Cologne, 50931Cologne, Germany
- Center for Molecular Medicine Cologne, University of Cologne, 50931Cologne, Germany
| | - Stefan Kohl
- Children’s and Adolescents’ Hospital, University Hospital Cologne, Faculty of Medicine, University of Cologne, 50937Cologne, Germany
| | - Nikolai Tschernoster
- Institute of Human Genetics, University Hospital Cologne, Faculty of Medicine, University of Cologne, 50931Cologne, Germany
- Center for Molecular Medicine Cologne, University of Cologne, 50931Cologne, Germany
- Cologne Center for Genomics, University of Cologne, 50931Cologne, Germany
| | - Gunter Rappl
- Center for Molecular Medicine Cologne, University of Cologne, 50931Cologne, Germany
| | - Isabelle Rouvet
- Centre de Biotechnologie Cellulaire and CBC BioTec Biobank, Centre de Ressources Biologiques, Hospices Civils de Lyon, 69229Lyon, France
| | | | - Felix S. Seibert
- Medical Department I, University Hospital Marien Hospital Herne, Ruhr-University Bochum, 44625Herne, Germany
| | - Holger Thiele
- Cologne Center for Genomics, University of Cologne, 50931Cologne, Germany
| | - Martin G. Häusler
- Division of Neuropediatrics and Social Pediatrics, Department of Pediatrics, Medical Faculty, Rheinisch-Westfälische Technische Hochschule Aachen University, 52074Aachen, Germany
| | - Lutz T. Weber
- Children’s and Adolescents’ Hospital, University Hospital Cologne, Faculty of Medicine, University of Cologne, 50937Cologne, Germany
| | - Maike Büttner-Herold
- Department of Nephropathology, Institute of Pathology, Friedrich-Alexander Universität Erlangen-Nürnberg (FAU), 91054Erlangen, Germany
| | - Miriam Elbracht
- Institute for Human Genetics and Genomic Medicine, Medical Faculty, Rheinisch-Westfälische Technische Hochschule Aachen University, 52074Aachen, Germany
| | - Sandra F. Cummings
- Division of Surgical Sciences, Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA02215
| | - Janine Altmüller
- Center for Molecular Medicine Cologne, University of Cologne, 50931Cologne, Germany
- Cologne Center for Genomics, University of Cologne, 50931Cologne, Germany
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Core Facility Genomics, 10178Berlin, Germany
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, 13125Berlin, Germany
| | - Sandra Habbig
- Children’s and Adolescents’ Hospital, University Hospital Cologne, Faculty of Medicine, University of Cologne, 50937Cologne, Germany
| | - Richard D. Cummings
- Division of Surgical Sciences, Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA02215
| | - Bodo B. Beck
- Institute of Human Genetics, University Hospital Cologne, Faculty of Medicine, University of Cologne, 50931Cologne, Germany
- Center for Molecular Medicine Cologne, University of Cologne, 50931Cologne, Germany
| |
Collapse
|
12
|
Lin J, Ehinger E, Hanna DB, Qi Q, Wang T, Ghosheh Y, Mueller K, Anastos K, Lazar JM, Mack WJ, Tien PC, Berman JW, Cohen MH, Ofotokun I, Gange S, Liu C, Heath SL, Tracy RP, Hodis HN, Landay AL, Ley K, Kaplan RC. HIV infection and cardiovascular disease have both shared and distinct monocyte gene expression features: Women's Interagency HIV study. PLoS One 2023; 18:e0285926. [PMID: 37205656 PMCID: PMC10198505 DOI: 10.1371/journal.pone.0285926] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2023] [Accepted: 05/04/2023] [Indexed: 05/21/2023] Open
Abstract
Persistent inflammation contributes to the development of cardiovascular disease (CVD) as an HIV-associated comorbidity. Innate immune cells such as monocytes are major drivers of inflammation in men and women with HIV. The study objectives are to examine the contribution of circulating non-classical monocytes (NCM, CD14dimCD16+) and intermediate monocytes (IM, CD14+CD16+) to the host response to long-term HIV infection and HIV-associated CVD. Women with and without chronic HIV infection (H) were studied. Subclinical CVD (C) was detected as plaques imaged by B-mode carotid artery ultrasound. The study included H-C-, H+C-, H-C+, and H+C+ participants (23 of each, matched on race/ethnicity, age and smoking status), selected from among enrollees in the Women's Interagency HIV Study. We assessed transcriptomic features associated with HIV or CVD alone or comorbid HIV/CVD comparing to healthy (H-C-) participants in IM and NCM isolated from peripheral blood mononuclear cells. IM gene expression was little affected by HIV alone or CVD alone. In IM, coexisting HIV and CVD produced a measurable gene transcription signature, which was abolished by lipid-lowering treatment. In NCM, versus non-HIV controls, women with HIV had altered gene expression, irrespective of whether or not they had comorbid CVD. The largest set of differentially expressed genes was found in NCM among women with both HIV and CVD. Genes upregulated in association with HIV included several potential targets of drug therapies, including LAG3 (CD223). In conclusion, circulating monocytes from patients with well controlled HIV infection demonstrate an extensive gene expression signature which may be consistent with the ability of these cells to serve as potential viral reservoirs. Gene transcriptional changes in HIV patients were further magnified in the presence of subclinical CVD.
Collapse
Affiliation(s)
- Juan Lin
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, NY, United States of America
| | - Erik Ehinger
- Department of Inflammation Biology, La Jolla Institute for Allergy and Immunology, La Jolla, CA, United States of America
| | - David B. Hanna
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, NY, United States of America
| | - Qibin Qi
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, NY, United States of America
| | - Tao Wang
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, NY, United States of America
| | - Yanal Ghosheh
- Department of Inflammation Biology, La Jolla Institute for Allergy and Immunology, La Jolla, CA, United States of America
| | - Karin Mueller
- Department of Cardiology, Eberhard Karls University, Tuebingen University Hospital, Tuebingen, Germany
| | - Kathryn Anastos
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, NY, United States of America
- Department of Medicine, Albert Einstein College of Medicine, Bronx, NY, United States of America
| | - Jason M. Lazar
- Department of Medicine, Downstate Medical Center, State University of New York, Brooklyn, NY, United States of America
| | - Wendy J. Mack
- Department of Preventive Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States of America
| | - Phyllis C. Tien
- Department of Medicine, and Department of Veterans Affairs, Medical Center, University of California, San Francisco, San Francisco, CA, United States of America
| | - Joan W. Berman
- Department of Pathology, Albert Einstein College of Medicine, Bronx, NY, United States of America
| | - Mardge H. Cohen
- Department of Medicine, John Stroger Hospital and Rush University, Chicago, IL, United States of America
| | - Igho Ofotokun
- Department of Medicine, Infectious Disease Division and Grady Health Care System, Emory University School of Medicine, Atlanta, GA, United States of America
| | - Stephen Gange
- Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, United States of America
| | - Chenglong Liu
- Department of Medicine, Georgetown University Medical Center, Washington, DC, United States of America
| | - Sonya L. Heath
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, United States of America
| | - Russell P. Tracy
- Department of Pathology & Laboratory Medicine and Biochemistry, University of Vermont Larner College of Medicine, Colchester, VT, United States of America
| | - Howard N. Hodis
- Department of Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States of America
| | - Alan L. Landay
- Department of Internal Medicine, Rush University Medical Center, Chicago, IL, United States of America
| | - Klaus Ley
- Department of Inflammation Biology, La Jolla Institute for Allergy and Immunology, La Jolla, CA, United States of America
- Department of Bioengineering, University of California San Diego, San Diego, CA, United States of America
| | - Robert C. Kaplan
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, NY, United States of America
- Fred Hutchinson Cancer Research Center, Division of Public Health Sciences, Seattle, WA, United States of America
| |
Collapse
|
13
|
Li J, Qiu Y, Zhang C, Wang H, Bi R, Wei Y, Li Y, Hu B. The role of protein glycosylation in the occurrence and outcome of acute ischemic stroke. Pharmacol Res 2023; 191:106726. [PMID: 36907285 DOI: 10.1016/j.phrs.2023.106726] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Revised: 03/03/2023] [Accepted: 03/09/2023] [Indexed: 03/12/2023]
Abstract
Acute ischemic stroke (AIS) is a serious and life-threatening disease worldwide. Despite thrombolysis or endovascular thrombectomy, a sizeable fraction of patients with AIS have adverse clinical outcomes. In addition, existing secondary prevention strategies with antiplatelet and anticoagulant drugs therapy are not able to adequately decrease the risk of ischemic stroke recurrence. Thus, exploring novel mechanisms for doing so represents an urgent need for the prevention and treatment of AIS. Recent studies have discovered that protein glycosylation plays a critical role in the occurrence and outcome of AIS. As a common co- and post-translational modification, protein glycosylation participates in a wide variety of physiological and pathological processes by regulating the activity and function of proteins or enzymes. Protein glycosylation is involved in two causes of cerebral emboli in ischemic stroke: atherosclerosis and atrial fibrillation. Following ischemic stroke, the level of brain protein glycosylation becomes dynamically regulated, which significantly affects stroke outcome through influencing inflammatory response, excitotoxicity, neuronal apoptosis, and blood-brain barrier disruption. Drugs targeting glycosylation in the occurrence and progression of stroke may represent a novel therapeutic idea. In this review, we focus on possible perspectives about how glycosylation affects the occurrence and outcome of AIS. We then propose the potential of glycosylation as a therapeutic drug target and prognostic marker for AIS patients in the future.
Collapse
Affiliation(s)
- Jianzhuang Li
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yanmei Qiu
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Chunlin Zhang
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hailing Wang
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Rentang Bi
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yanhao Wei
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yanan Li
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Bo Hu
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| |
Collapse
|
14
|
A role for endothelial alpha-mannosidase MAN1C1 in radiation-induced immune cell recruitment. iScience 2022; 25:105482. [DOI: 10.1016/j.isci.2022.105482] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Revised: 10/06/2022] [Accepted: 10/28/2022] [Indexed: 11/15/2022] Open
|
15
|
Effects of Highly Pathogenic Porcine Reproductive and Respiratory Syndrome Virus Infection on the Surface Glycoprofiling of Porcine Pulmonary Microvascular Endothelial Cells. Viruses 2022; 14:v14112569. [PMID: 36423178 PMCID: PMC9695484 DOI: 10.3390/v14112569] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Revised: 11/18/2022] [Accepted: 11/18/2022] [Indexed: 11/22/2022] Open
Abstract
Previously, our study has demonstrated that porcine pulmonary microvascular endothelial cells (PPMVECs) were susceptible to highly pathogenic porcine reproductive and respiratory syndrome virus (HP-PRRSV) and produced a significant non-specific immune response to it. The significance of microvascular endothelial glycocalyx is increasingly attracting attention, and its rich carbohydrate components are not only important signaling molecules, but also remarkably influence the signaling of most proteins. Comprehending changes in the carbohydrate chains contributes to understanding cell functions. This study aimed to reveal the effects of HP-PRRSV infection on the surface carbohydrate chains of PPMVECs. PPMVECs were isolated and cultured in vitro and infected with HP-PRRSV HN and JXA1 strains. Scanning electron microscopy analysis indicated that at 48 h post-infection, some broken holes were in their cell membranes, and that the surface fibrous glycocalyx was obviously reduced or even disappeared. Lectin microarray analysis indicated that the fluorescence intensities of 8 and 7 lectin sites were significantly changed by the HP-PRRSV HN and JXA1 strains, respectively, among which there were 6 common lectin sites. The up-regulation of common lectins (RCA-I, LEL, and STL) and the down-regulation of common lectins (LCA, DSA, and PHA-E) were confirmed by lectin fluorescence staining and lectin flow cytometry, respectively. Together, the results show that the HP-PRRSV infection can induce the glycocalyx disruption of PPMVECs and their surface glycoprofiling changes, and that the poly-N-acetyllactosamine and complex N-glycan are the main up-regulated and down-regulated carbohydrate chains, respectively. Our findings may provide insights into revealing the pathogenesis of HP-PRRSV from the perspective of glycobiology.
Collapse
|
16
|
Radovani B, Gudelj I. N-Glycosylation and Inflammation; the Not-So-Sweet Relation. Front Immunol 2022; 13:893365. [PMID: 35833138 PMCID: PMC9272703 DOI: 10.3389/fimmu.2022.893365] [Citation(s) in RCA: 61] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Accepted: 05/30/2022] [Indexed: 12/28/2022] Open
Abstract
Chronic inflammation is the main feature of many long-term inflammatory diseases such as autoimmune diseases, metabolic disorders, and cancer. There is a growing number of studies in which alterations of N-glycosylation have been observed in many pathophysiological conditions, yet studies of the underlying mechanisms that precede N-glycome changes are still sparse. Proinflammatory cytokines have been shown to alter the substrate synthesis pathways as well as the expression of glycosyltransferases required for the biosynthesis of N-glycans. The resulting N-glycosylation changes can further contribute to disease pathogenesis through modulation of various aspects of immune cell processes, including those relevant to pathogen recognition and fine-tuning the inflammatory response. This review summarizes our current knowledge of inflammation-induced N-glycosylation changes, with a particular focus on specific subsets of immune cells of innate and adaptive immunity and how these changes affect their effector functions, cell interactions, and signal transduction.
Collapse
Affiliation(s)
- Barbara Radovani
- Department of Biotechnology, University of Rijeka, Rijeka, Croatia
| | - Ivan Gudelj
- Department of Biotechnology, University of Rijeka, Rijeka, Croatia
- Genos Glycoscience Research Laboratory, Zagreb, Croatia
| |
Collapse
|
17
|
Tang Z, Sarnat JA, Weber RJ, Russell AG, Zhang X, Li Z, Yu T, Jones DP, Liang D. The Oxidative Potential of Fine Particulate Matter and Biological Perturbations in Human Plasma and Saliva Metabolome. ENVIRONMENTAL SCIENCE & TECHNOLOGY 2022; 56:7350-7361. [PMID: 35075906 PMCID: PMC9177558 DOI: 10.1021/acs.est.1c04915] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/03/2023]
Abstract
Particulate oxidative potential may comprise a key health-relevant parameter of particulate matter (PM) toxicity. To identify biological perturbations associated with particulate oxidative potential and examine the underlying molecular mechanisms, we recruited 54 participants from two dormitories near and far from a congested highway in Atlanta, GA. Fine particulate matter oxidative potential ("FPMOP") levels at the dormitories were measured using dithiothreitol assay. Plasma and saliva samples were collected from participants four times for longitudinal high-resolution metabolic profiling. We conducted metabolome-wide association studies to identify metabolic signals with FPMOP. Leukotriene metabolism and galactose metabolism were top pathways associated with ≥5 FPMOP-related indicators in plasma, while vitamin E metabolism and leukotriene metabolism were found associated with most FPMOP indicators in saliva. We observed different patterns of perturbed pathways significantly associated with water-soluble and -insoluble FPMOPs, respectively. We confirmed five metabolites directly associated with FPMOP, including hypoxanthine, histidine, pyruvate, lactate/glyceraldehyde, and azelaic acid, which were implications of perturbations in acute inflammation, nucleic acid damage and repair, and energy perturbation. The unique metabolic signals were specific to FPMOP, but not PM mass, providing initial indication that FPMOP might constitute a more sensitive, health-relevant measure for elucidating etiologies related to PM2.5 exposures.
Collapse
Affiliation(s)
- Ziyin Tang
- Gangarosa Department of Environmental Health, Rollins School of Public Health, Emory University, Atlanta, Georgia 30322, United States
| | - Jeremy A Sarnat
- Gangarosa Department of Environmental Health, Rollins School of Public Health, Emory University, Atlanta, Georgia 30322, United States
| | - Rodney J Weber
- School of Earth and Atmospheric Sciences, Georgia Institute of Technology, Atlanta, Georgia 30322, United States
| | - Armistead G Russell
- School of Civil and Environmental Engineering, Georgia Institute of Technology, Atlanta, Georgia 30322, United States
| | - Xiaoyue Zhang
- Gangarosa Department of Environmental Health, Rollins School of Public Health, Emory University, Atlanta, Georgia 30322, United States
| | - Zhenjiang Li
- Gangarosa Department of Environmental Health, Rollins School of Public Health, Emory University, Atlanta, Georgia 30322, United States
| | - Tianwei Yu
- School of Data Science, The Chinese University of Hong Kong, Shenzhen, Guangdong 518172, China
| | - Dean P Jones
- Department of Medicine, School of Medicine, Emory University, Atlanta, Georgia 30322, United States
| | - Donghai Liang
- Gangarosa Department of Environmental Health, Rollins School of Public Health, Emory University, Atlanta, Georgia 30322, United States
| |
Collapse
|
18
|
Heindel D, Chen S, Aziz PV, Chung JY, Marth JD, Mahal LK. Glycomic Analysis Reveals a Conserved Response to Bacterial Sepsis Induced by Different Bacterial Pathogens. ACS Infect Dis 2022; 8:1075-1085. [PMID: 35486714 PMCID: PMC9112329 DOI: 10.1021/acsinfecdis.2c00082] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Indexed: 12/15/2022]
Abstract
Sepsis is an extreme inflammatory response to infection that occurs in the bloodstream and causes damage throughout the body. Glycosylation is known to play a role in immunity and inflammation, but the role of glycans in sepsis is not well-defined. Herein, we profiled the serum glycomes of experimental mouse sepsis models to identify changes induced by 4 different clinical bacterial pathogens (Gram-positive: Streptococcus pneumoniae and Staphylococcus aureus, Gram-negative: Escherichia coli and Salmonella Typhimurium) using our lectin microarray technology. We observed global shifts in the blood sera glycome that were conserved across all four species, regardless of whether they were Gram positive or negative. Bisecting GlcNAc was decreased upon sepsis and a strong increase in core 1/3 O-glycans was observed. Lectin blot analysis revealed a high molecular weight protein induced in sepsis by all four bacteria as the major cause of the core 1/3 O-glycan shift. Analysis of this band by mass spectrometry identified interalpha-trypsin inhibitor heavy chains (ITIHs) and fibronectin, both of which are associated with human sepsis. Shifts in the glycosylation of these proteins were observed. Overall, our work points toward a common mechanism for bacterially induced sepsis, marked by conserved changes in the glycome.
Collapse
Affiliation(s)
- Daniel
W. Heindel
- Biomedical
Research Institute, Department of Chemistry, New York University, New York, New York 10003, United States
| | - Shuhui Chen
- Biomedical
Research Institute, Department of Chemistry, New York University, New York, New York 10003, United States
| | - Peter V. Aziz
- SBP
Medical Discovery Institute, La Jolla, California 92037, United States
| | - Jonathan Y. Chung
- Biomedical
Research Institute, Department of Chemistry, New York University, New York, New York 10003, United States
| | - Jamey D. Marth
- SBP
Medical Discovery Institute, La Jolla, California 92037, United States
| | - Lara K. Mahal
- Biomedical
Research Institute, Department of Chemistry, New York University, New York, New York 10003, United States
- Department
of Chemistry, University of Alberta, Edmonton, AB, T6G 2G2, Canada
| |
Collapse
|
19
|
He L, Chung PHY, Lui VCH, Tang CSM, Tam PKH. Current Understanding in the Clinical Characteristics and Molecular Mechanisms in Different Subtypes of Biliary Atresia. Int J Mol Sci 2022; 23:ijms23094841. [PMID: 35563229 PMCID: PMC9103665 DOI: 10.3390/ijms23094841] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Revised: 04/23/2022] [Accepted: 04/24/2022] [Indexed: 02/01/2023] Open
Abstract
Biliary atresia is a severe obliterative cholangiopathy in early infancy that is by far the most common cause of surgical jaundice and the most common indicator for liver transplantation in children. With the advanced knowledge gained from different clinical trials and the development of research models, a more precise clinical classification of BA (i.e., isolated BA (IBA), cystic BA (CBA), syndromic BA (SBA), and cytomegalovirus-associated BA (CMVBA)) is proposed. Different BA subtypes have similar yet distinguishable clinical manifestations. The clinical and etiological heterogeneity leads to dramatically different prognoses; hence, treatment needs to be specific. In this study, we reviewed the clinical characteristics of different BA subtypes and revealed the molecular mechanisms of their developmental contributors. We aimed to highlight the differences among these various subtypes of BA which ultimately contribute to the development of a specific management protocol for each subtype.
Collapse
Affiliation(s)
- Lin He
- Cancer Centre, Faculty of Health Sciences, University of Macau, Macau SAR, China;
| | - Patrick Ho Yu Chung
- Division of Paediatric Surgery, Department of Surgery, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China; (V.C.H.L.); (C.S.M.T.); (P.K.H.T.)
- Correspondence: ; Tel.: +852-22554850; Fax: +852-28173155
| | - Vincent Chi Hang Lui
- Division of Paediatric Surgery, Department of Surgery, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China; (V.C.H.L.); (C.S.M.T.); (P.K.H.T.)
| | - Clara Sze Man Tang
- Division of Paediatric Surgery, Department of Surgery, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China; (V.C.H.L.); (C.S.M.T.); (P.K.H.T.)
| | - Paul Kwong Hang Tam
- Division of Paediatric Surgery, Department of Surgery, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China; (V.C.H.L.); (C.S.M.T.); (P.K.H.T.)
- Faculty of Medicine, Macau University of Science and Technology, Macau SAR, China
| |
Collapse
|
20
|
Kumar D, Sharma SR, Adegoke A, Kennedy A, Tuten HC, Li AY, Karim S. Recently Evolved Francisella-Like Endosymbiont Outcompetes an Ancient and Evolutionarily Associated Coxiella-Like Endosymbiont in the Lone Star Tick ( Amblyomma americanum) Linked to the Alpha-Gal Syndrome. Front Cell Infect Microbiol 2022; 12:787209. [PMID: 35493735 PMCID: PMC9039623 DOI: 10.3389/fcimb.2022.787209] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Accepted: 03/18/2022] [Indexed: 12/04/2022] Open
Abstract
Background Ticks are hematophagous arthropods that transmit various bacterial, viral, and protozoan pathogens of public health significance. The lone star tick (Amblyomma americanum) is an aggressive human-biting tick that transmits bacterial and viral pathogens, and its bites are suspected of eliciting the alpha-gal syndrome, a newly emerged delayed hypersensitivity following consumption of red meat in the United States. While ongoing studies have attempted to investigate the contribution of different tick-inherent factors to the induction of alpha-gal syndrome, an otherwise understudied aspect is the contribution of the tick microbiome and specifically obligate endosymbionts to the establishment of the alpha-gal syndrome in humans. Materials and Methods Here we utilized a high-throughput metagenomic sequencing approach to cataloging the entire microbial communities residing within different developmental stages and tissues of unfed and blood-fed ticks from laboratory-maintained ticks and three new geographical locations in the United States. The Quantitative Insights Into Microbial Ecology (QIIME2) pipeline was used to perform data analysis and taxonomic classification. Moreover, using a SparCC (Sparse Correlations for Compositional data) network construction model, we investigated potential interactions between members of the microbial communities from laboratory-maintained and field-collected ticks. Results Overall, Francisellaceae was the most dominant bacteria identified in the microbiome of both laboratory-raised and field-collected Am. americanum across all tissues and developmental stages. Likewise, microbial diversity was seen to be significantly higher in field-collected ticks compared with laboratory-maintained ticks as seen with a higher number of both Operational Taxonomic Units and measures of species richness. Several potential positive and negative correlations were identified from our network analysis. We observed a strong positive correlation between Francisellaceae, Rickettsiaceae, and Midichloriaceae in both developmental stages and tissues from laboratory-maintained ticks, whereas ovarian tissues had a strong positive correlation of bacteria in the family Xanthobacteraceae and Rhizobiaceae. A negative interaction was observed between Coxiellaceae and Francisellaceae in Illinois, and all the bacteria detected from ticks from Delaware were negatively correlated. Conclusion This study is the first to catalog the microbiome of Am. americanum throughout its developmental stages and different tissue niches and report the potential replacement of Coxiellaceae by Francisellaceae across developmental stages and tissues tested except in ovarian tissues. These unique and significant findings advance our knowledge and open a new avenue of research to further understand the role of tick microbiome in tick-borne diseases and develop a holistic strategy to control alpha-gal syndrome.
Collapse
Affiliation(s)
- Deepak Kumar
- School of Biological, Environmental, and Earth Sciences, University of Southern Mississippi, Hattiesburg, MS, United States
| | - Surendra Raj Sharma
- School of Biological, Environmental, and Earth Sciences, University of Southern Mississippi, Hattiesburg, MS, United States
| | - Abdulsalam Adegoke
- School of Biological, Environmental, and Earth Sciences, University of Southern Mississippi, Hattiesburg, MS, United States
| | - Ashley Kennedy
- Delaware Division of Fish & Wildlife, Delaware Mosquito Control Sect., Newark, DE, United States
| | - Holly C. Tuten
- Illinois Natural History Survey (INHS), University of Illinois Urbana-Champaign, Champaign, IL, United States
| | - Andrew Y. Li
- Invasive Insect Biocontrol & Behavior Laboratory, United States Department of Agriculture, Agricultural Research Service (USDA ARS), Beltsville, MD, United States
| | - Shahid Karim
- School of Biological, Environmental, and Earth Sciences, University of Southern Mississippi, Hattiesburg, MS, United States
- Center for Molecular and Cellular Biosciences, University of Southern Mississippi, Hattiesburg, MS, United States
| |
Collapse
|
21
|
Rajagopal MU, Bansal S, Kaur P, Jain SK, Altadil T, Hinzman CP, Li Y, Moulton J, Singh B, Bansal S, Chauthe SK, Singh R, Banerjee PP, Mapstone M, Fiandaca MS, Federoff HJ, Unger K, Smith JP, Cheema AK. TGFβ Drives Metabolic Perturbations during Epithelial Mesenchymal Transition in Pancreatic Cancer: TGFβ Induced EMT in PDAC. Cancers (Basel) 2021; 13:cancers13246204. [PMID: 34944824 PMCID: PMC8699757 DOI: 10.3390/cancers13246204] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Revised: 11/26/2021] [Accepted: 12/06/2021] [Indexed: 11/16/2022] Open
Abstract
Simple Summary Pancreatic cancer is an aggressive disease with most patients diagnosed at late stages resulting in poor outcomes. While it is known that pancreatic tumor cells undergo epithelial to mesenchymal transition, the metabolic alterations accompanying that transition are not characterized. This study leveraged a metabolomics approach to understand the small molecule and biochemical perturbations that can be targeted for designing strategies for improving outcomes in pancreatic cancer. Abstract Pancreatic ductal adenocarcinoma (PDAC) is a highly lethal malignancy wherein a majority of patients present metastatic disease at diagnosis. Although the role of epithelial to mesenchymal transition (EMT), mediated by transforming growth factor beta (TGFβ), in imparting an aggressive phenotype to PDAC is well documented, the underlying biochemical pathway perturbations driving this behaviour have not been elucidated. We used high-resolution mass spectrometry (HRMS) based molecular phenotyping approach in order to delineate metabolic changes concomitant to TGFβ-induced EMT in pancreatic cancer cells. Strikingly, we observed robust changes in amino acid and energy metabolism that may contribute to tumor invasion and metastasis. Somewhat unexpectedly, TGFβ treatment resulted in an increase in intracellular levels of retinoic acid (RA) that in turn resulted in increased levels of extracellular matrix (ECM) proteins including fibronectin (FN) and collagen (COL1). These findings were further validated in plasma samples obtained from patients with resectable pancreatic cancer. Taken together, these observations provide novel insights into small molecule dysregulation that triggers a molecular cascade resulting in increased EMT-like changes in pancreatic cancer cells, a paradigm that can be potentially targeted for better clinical outcomes.
Collapse
Affiliation(s)
- Meena U. Rajagopal
- Department of Oncology, Georgetown University Medical Center, Washington, DC 20057, USA; (M.U.R.); (S.B.); (Y.L.); (J.M.); (B.S.); (S.B.)
| | - Shivani Bansal
- Department of Oncology, Georgetown University Medical Center, Washington, DC 20057, USA; (M.U.R.); (S.B.); (Y.L.); (J.M.); (B.S.); (S.B.)
| | - Prabhjit Kaur
- Department of Botany, Khalsa College, Amritsar 143002, India; (P.K.); (R.S.)
| | - Shreyans K. Jain
- Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology, Banaras Hindu University, Varanasi 221005, India;
| | - Tatiana Altadil
- Biomedical Research Group in Gynaecology, Vall Hebron Research Institute (VHIR), Universitat Autònoma de Barcelona, 08035 Barcelona, Spain;
| | - Charles P. Hinzman
- Department of Biochemistry and Molecular and Cellular Biology, Georgetown University Medical Center, Washington, DC 20057, USA; (C.P.H.); (P.P.B.)
| | - Yaoxiang Li
- Department of Oncology, Georgetown University Medical Center, Washington, DC 20057, USA; (M.U.R.); (S.B.); (Y.L.); (J.M.); (B.S.); (S.B.)
| | - Joanna Moulton
- Department of Oncology, Georgetown University Medical Center, Washington, DC 20057, USA; (M.U.R.); (S.B.); (Y.L.); (J.M.); (B.S.); (S.B.)
| | - Baldev Singh
- Department of Oncology, Georgetown University Medical Center, Washington, DC 20057, USA; (M.U.R.); (S.B.); (Y.L.); (J.M.); (B.S.); (S.B.)
| | - Sunil Bansal
- Department of Oncology, Georgetown University Medical Center, Washington, DC 20057, USA; (M.U.R.); (S.B.); (Y.L.); (J.M.); (B.S.); (S.B.)
| | - Siddheshwar Kisan Chauthe
- Department of Natural Products, National Institute of Pharmaceutical Education and Research (NIPER), Ahmedabad 380054, India;
| | - Rajbir Singh
- Department of Botany, Khalsa College, Amritsar 143002, India; (P.K.); (R.S.)
| | - Partha P. Banerjee
- Department of Biochemistry and Molecular and Cellular Biology, Georgetown University Medical Center, Washington, DC 20057, USA; (C.P.H.); (P.P.B.)
| | - Mark Mapstone
- Department of Neurology, University of California, Irvine, CA 92697, USA; (M.M.); (M.S.F.); (H.J.F.)
| | - Massimo S. Fiandaca
- Department of Neurology, University of California, Irvine, CA 92697, USA; (M.M.); (M.S.F.); (H.J.F.)
- Department of Neurological Surgery, University of California, Irvine, CA 92697, USA
| | - Howard J. Federoff
- Department of Neurology, University of California, Irvine, CA 92697, USA; (M.M.); (M.S.F.); (H.J.F.)
| | - Keith Unger
- Radiation Medicine, Med-Star Georgetown University Hospital, Washington, DC 20057, USA;
| | - Jill P. Smith
- Department of Medicine, Georgetown University Medical Center, Washington, DC 20057, USA;
| | - Amrita K. Cheema
- Department of Oncology, Georgetown University Medical Center, Washington, DC 20057, USA; (M.U.R.); (S.B.); (Y.L.); (J.M.); (B.S.); (S.B.)
- Department of Biochemistry and Molecular and Cellular Biology, Georgetown University Medical Center, Washington, DC 20057, USA; (C.P.H.); (P.P.B.)
- Correspondence: ; Tel.: +1-202-687-2756; Fax: +1-202-687-8860
| |
Collapse
|
22
|
Nazemi SA, Olesińska M, Pezzella C, Varriale S, Lin CW, Corvini PFX, Shahgaldian P. Immobilisation and stabilisation of glycosylated enzymes on boronic acid-functionalised silica nanoparticles. Chem Commun (Camb) 2021; 57:11960-11963. [PMID: 34705002 DOI: 10.1039/d1cc04916j] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
We report a method of glycosylated enzymes' surface immobilisation and stabilisation. The enzyme is immobilised at the surface of silica nanoparticles through the reversible covalent binding of vicinal diols of the enzyme glycans with a surface-attached boronate derivative. A soft organosilica layer of controlled thickness is grown at the silica surface, entrapping the enzyme and thus avoiding enzyme leaching. We demonstrate that this approach results not only in high and durable activity retention but also enzyme stabilisation.
Collapse
Affiliation(s)
- Seyed Amirabbas Nazemi
- School of Life Science, University of Applied Sciences and Arts Northwestern Switzerland, Hofackerstrasee 30, Muttenz CH-4132, Switzerland.
| | - Magdalena Olesińska
- School of Life Science, University of Applied Sciences and Arts Northwestern Switzerland, Hofackerstrasee 30, Muttenz CH-4132, Switzerland.
| | - Cinzia Pezzella
- Biopox, Viale Maria Bakunin, 12 - CAP 80125 Naples, Italy.,Department of Agricultural Sciences, University of Naples Federico II, Via Università, 100 80055 Portici, NA, Italy
| | | | - Chia-Wei Lin
- Functional Genomics Center Zürich, University of Zürich/ETH Zürich, 8057 Zürich, Switzerland
| | - Philippe F-X Corvini
- School of Life Science, University of Applied Sciences and Arts Northwestern Switzerland, Hofackerstrasee 30, Muttenz CH-4132, Switzerland.
| | - Patrick Shahgaldian
- School of Life Science, University of Applied Sciences and Arts Northwestern Switzerland, Hofackerstrasee 30, Muttenz CH-4132, Switzerland.
| |
Collapse
|
23
|
Trastoy B, Du JJ, Li C, García-Alija M, Klontz EH, Roberts BR, Donahue TC, Wang LX, Sundberg EJ, Guerin ME. GH18 endo-β-N-acetylglucosaminidases use distinct mechanisms to process hybrid-type N-linked glycans. J Biol Chem 2021; 297:101011. [PMID: 34324829 PMCID: PMC8374693 DOI: 10.1016/j.jbc.2021.101011] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Revised: 07/22/2021] [Accepted: 07/23/2021] [Indexed: 12/24/2022] Open
Abstract
N-glycosylation is one of the most abundant posttranslational modifications of proteins, essential for many physiological processes, including protein folding, protein stability, oligomerization and aggregation, and molecular recognition events. Defects in the N-glycosylation pathway cause diseases that are classified as congenital disorders of glycosylation. The ability to manipulate protein N-glycosylation is critical not only to our fundamental understanding of biology but also for the development of new drugs for a wide range of human diseases. Chemoenzymatic synthesis using engineered endo-β-N-acetylglucosaminidases (ENGases) has been used extensively to modulate the chemistry of N-glycosylated proteins. However, defining the molecular mechanisms by which ENGases specifically recognize and process N-glycans remains a major challenge. Here we present the X-ray crystal structure of the ENGase EndoBT-3987 from Bacteroides thetaiotaomicron in complex with a hybrid-type glycan product. In combination with alanine scanning mutagenesis, molecular docking calculations and enzymatic activity measurements conducted on a chemically engineered monoclonal antibody substrate unveil two mechanisms for hybrid-type recognition and processing by paradigmatic ENGases. Altogether, the experimental data provide pivotal insight into the molecular mechanism of substrate recognition and specificity for GH18 ENGases and further advance our understanding of chemoenzymatic synthesis and remodeling of homogeneous N-glycan glycoproteins.
Collapse
Affiliation(s)
- Beatriz Trastoy
- Structural Glycobiology Lab, Structural Biology Unit, Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Bizkaia, Derio, Spain; Structural Glycobiology Lab, IIS-Biocruces Bizkaia, Barakaldo, Bizkaia, Spain.
| | - Jonathan J Du
- Department of Biochemistry, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Chao Li
- Department of Chemistry and Biochemistry, University of Maryland, College Park, Maryland, USA
| | - Mikel García-Alija
- Structural Glycobiology Lab, Structural Biology Unit, Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Bizkaia, Derio, Spain; Structural Glycobiology Lab, IIS-Biocruces Bizkaia, Barakaldo, Bizkaia, Spain
| | - Erik H Klontz
- Institute of Human Virology, University of Maryland School of Medicine, Baltimore, Maryland, USA; Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Blaine R Roberts
- Department of Biochemistry, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Thomas C Donahue
- Department of Chemistry and Biochemistry, University of Maryland, College Park, Maryland, USA
| | - Lai-Xi Wang
- Department of Chemistry and Biochemistry, University of Maryland, College Park, Maryland, USA
| | - Eric J Sundberg
- Department of Biochemistry, Emory University School of Medicine, Atlanta, Georgia, USA.
| | - Marcelo E Guerin
- Structural Glycobiology Lab, Structural Biology Unit, Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Bizkaia, Derio, Spain; Structural Glycobiology Lab, IIS-Biocruces Bizkaia, Barakaldo, Bizkaia, Spain; Ikerbasque, Basque Foundation for Science, Bilbao, Spain.
| |
Collapse
|
24
|
Krick S, Helton ES, Easter M, Bollenbecker S, Denson R, Zaharias R, Cochran P, Vang S, Harris E, Wells JM, Barnes JW. ST6GAL1 and α2-6 Sialylation Regulates IL-6 Expression and Secretion in Chronic Obstructive Pulmonary Disease. Front Immunol 2021; 12:693149. [PMID: 34290711 PMCID: PMC8287524 DOI: 10.3389/fimmu.2021.693149] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Accepted: 06/21/2021] [Indexed: 01/23/2023] Open
Abstract
Chronic obstructive pulmonary disease (COPD) is a systemic disease strongly associated with cigarette smoking, airway inflammation, and acute disease exacerbations. Changes in terminal sialylation and fucosylation of asparagine (N)-linked glycans have been documented in COPD, but the role that glycosyltransferases may play in the regulation of N-linked glycans in COPD has not been fully elucidated. Recent studies suggest that modulation of ST6GAL1 (ST6 beta-galactoside alpha-2,6-sialyltransferase-1), which catalyzes terminal α2-6 sialylation of cellular proteins, may regulate inflammation and contribute to COPD phenotype(s). Interestingly, it has been previously demonstrated that ST6GAL1, a Golgi resident protein, can be proteolytically processed by BACE1 (beta-site amyloid precursor protein cleaving enzyme-1) to a circulating form that retains activity. In this study, we showed that loss of ST6GAL1 expression increased interleukin (IL)-6 expression and secretion in human bronchial epithelial cells (HBECs). Furthermore, exposure to cigarette smoke medium/extract (CSE) or BACE1 inhibition resulted in decreased ST6GAL1 secretion, reduced α2-6 sialylation, and increased IL-6 production in HBECs. Analysis of plasma ST6GAL1 levels in a small COPD patient cohort demonstrated an inverse association with prospective acute exacerbations of COPD (AECOPD), while IL-6 was positively associated. Altogether, these results suggest that reduced ST6GAL1 and α2-6 sialylation augments IL-6 expression/secretion in HBECs and is associated with poor clinical outcomes in COPD.
Collapse
Affiliation(s)
- Stefanie Krick
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, The University of Alabama at Birmingham, Birmingham, AL, United States
- Gregory Fleming James Cystic Fibrosis Research Center, The University of Alabama at Birmingham, Birmingham, AL, United States
| | - E. Scott Helton
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, The University of Alabama at Birmingham, Birmingham, AL, United States
| | - Molly Easter
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, The University of Alabama at Birmingham, Birmingham, AL, United States
| | - Seth Bollenbecker
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, The University of Alabama at Birmingham, Birmingham, AL, United States
| | - Rebecca Denson
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, The University of Alabama at Birmingham, Birmingham, AL, United States
| | - Rennan Zaharias
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, The University of Alabama at Birmingham, Birmingham, AL, United States
| | - Phillip Cochran
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, The University of Alabama at Birmingham, Birmingham, AL, United States
| | - Shia Vang
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, The University of Alabama at Birmingham, Birmingham, AL, United States
| | - Elex Harris
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, The University of Alabama at Birmingham, Birmingham, AL, United States
- Gregory Fleming James Cystic Fibrosis Research Center, The University of Alabama at Birmingham, Birmingham, AL, United States
| | - James M. Wells
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, The University of Alabama at Birmingham, Birmingham, AL, United States
- UAB Lung Health Center, Birmingham, AL, United States
- Birmingham VA Medical Center, Birmingham, AL, United States
| | - Jarrod W. Barnes
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, The University of Alabama at Birmingham, Birmingham, AL, United States
- Gregory Fleming James Cystic Fibrosis Research Center, The University of Alabama at Birmingham, Birmingham, AL, United States
| |
Collapse
|
25
|
Bai Z, Xu L, Dai Y, Yuan Q, Zhou Z. ECM2 and GLT8D2 in human pulmonary artery hypertension: fruits from weighted gene co-expression network analysis. J Thorac Dis 2021; 13:2242-2254. [PMID: 34012575 PMCID: PMC8107565 DOI: 10.21037/jtd-20-3069] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Background Pulmonary artery hypertension (PAH) is an incurable disease with a high mortality rate. Current medications ameliorate symptoms but cannot target adverse vascular remodeling. New therapeutic strategies for PAH need to be established. Methods Using the weighted gene coexpression network analysis (WGCNA) algorithm, we constructed a coexpression network of dataset GSE117261 from the Gene Expression Omnibus (GEO) database. Key modules were identified by the relationship between module eigengenes and clinical traits. Hub genes were screened out based on gene significance (GS), module membership (MM), and mean pulmonary artery pressure (mPAP). External validations were conducted in GSE48149 and GSE113439. Functional enrichment and immune cell infiltration were analyzed using Metascape and CIBERSORTx. Results The WGCNA analysis revealed 13 coexpression modules. The pink module had the highest correlation with PAH in terms of module eigengene (r=0.79; P=2e-18) and module significance (MS =0.43). Functional enrichment indicated genes in the pink module contributed to the immune system process and extracellular matrix (ECM). In the pink module, ECM2 (GS =0.65, MM =0.86, ρ=0.407, P=0.0019) and GLT8D2 (GS =0.63, MM =0.85, ρ=0.443, P=0.006) were identified as hub genes. For immune cells infiltration in PAH lung tissue, hub genes were positively correlated with M2 macrophages and resting mast cells, and were negatively correlated with monocytes, neutrophils, and CD4-naïve T cells. Conclusions Our research identified 2 hub genes ECM2 and GLT8D2 related to PAH. The functions of these hub genes were involved in the immune process and ECM, indicating that they might serve as candidate therapeutic targets for PAH.
Collapse
Affiliation(s)
- Zeyang Bai
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Key Lab of Molecular Biological Targeted Therapies of the Ministry of Education, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Lianyan Xu
- Department of Radiology, Peking Union Medical College Hospital, PUMC & CAMS, Beijing, China
| | - Yong Dai
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Key Lab of Molecular Biological Targeted Therapies of the Ministry of Education, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Qingchen Yuan
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Key Lab of Molecular Biological Targeted Therapies of the Ministry of Education, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zihua Zhou
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Key Lab of Molecular Biological Targeted Therapies of the Ministry of Education, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
26
|
Krautter F, Iqbal AJ. Glycans and Glycan-Binding Proteins as Regulators and Potential Targets in Leukocyte Recruitment. Front Cell Dev Biol 2021; 9:624082. [PMID: 33614653 PMCID: PMC7890243 DOI: 10.3389/fcell.2021.624082] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Accepted: 01/12/2021] [Indexed: 12/19/2022] Open
Abstract
Leukocyte recruitment is a highly controlled cascade of interactions between proteins expressed by the endothelium and circulating leukocytes. The involvement of glycans and glycan-binding proteins in the leukocyte recruitment cascade has been well-characterised. However, our understanding of these interactions and their regulation has expanded substantially in recent years to include novel lectins and regulatory pathways. In this review, we discuss the role of glycans and glycan-binding proteins, mediating the interactions between endothelium and leukocytes both directly and indirectly. We also highlight recent findings of key enzymes involved in glycosylation which affect leukocyte recruitment. Finally, we investigate the potential of glycans and glycan binding proteins as therapeutic targets to modulate leukocyte recruitment and transmigration in inflammation.
Collapse
Affiliation(s)
- Franziska Krautter
- Institute of Cardiovascular Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Asif J Iqbal
- Institute of Cardiovascular Sciences, University of Birmingham, Birmingham, United Kingdom
| |
Collapse
|
27
|
Marsico G, Jin C, Abbah SA, Brauchle EM, Thomas D, Rebelo AL, Orbanić D, Chantepie S, Contessotto P, Papy-Garcia D, Rodriguez-Cabello C, Kilcoyne M, Schenke-Layland K, Karlsson NG, McCullagh KJA, Pandit A. Elastin-like hydrogel stimulates angiogenesis in a severe model of critical limb ischemia (CLI): An insight into the glyco-host response. Biomaterials 2021; 269:120641. [PMID: 33493768 DOI: 10.1016/j.biomaterials.2020.120641] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2020] [Revised: 12/24/2020] [Accepted: 12/29/2020] [Indexed: 12/15/2022]
Abstract
Critical limb ischemia (CLI) is characterized by the impairment of microcirculation, necrosis and inflammation of the muscular tissue. Although the role of glycans in mediating inflammation has been reported, changes in the glycosylation following muscle ischemia remains poorly understood. Here, a murine CLI model was used to show the increase of high mannose, α-(2, 6)-sialic acid and the decrease of hybrid and bisected N-glycans as glycosylation associated with the ischemic environment. Using this model, the efficacy of an elastin-like recombinamers (ELR) hydrogel was assessed. The hydrogel modulates key angiogenic signaling pathways, resulting in capillary formation, and ECM remodeling. Arterioles formation, reduction of fibrosis and anti-inflammatory macrophage polarization wa also induced by the hydrogel administration. Modulation of glycosylation was observed, suggesting, in particular, a role for mannosylation and sialylation in the mediation of tissue repair. Our study elucidates the angiogenic potential of the ELR hydrogel for CLI applications and identifies glycosylation alterations as potential new therapeutic targets.
Collapse
Affiliation(s)
- Grazia Marsico
- CÚRAM SFI Research Centre for Medical Devices, National University of Ireland Galway, Galway H92 W2TY, Ireland
| | - Chunseng Jin
- Department of Medical Biochemistry and Cell Biology at Institute of Biomedicine, Sahlgrenska Academy, The University of Gothenburg, Sweden
| | - Sunny A Abbah
- CÚRAM SFI Research Centre for Medical Devices, National University of Ireland Galway, Galway H92 W2TY, Ireland
| | - Eva M Brauchle
- Department of Women's Health, Research Institute for Women's Health, The Eberhard-Karls-University Tuebingen, Germany; The Natural and Medical Sciences Institute (NMI) at the University of Tübingen, Reutlingen, Germany
| | - Dilip Thomas
- CÚRAM SFI Research Centre for Medical Devices, National University of Ireland Galway, Galway H92 W2TY, Ireland
| | - Ana Lúcia Rebelo
- CÚRAM SFI Research Centre for Medical Devices, National University of Ireland Galway, Galway H92 W2TY, Ireland
| | | | - Sandrine Chantepie
- Cell Growth, Tissue Repair and Regeneration (CRRET), UPEC EA 4397/ERL CNRS 9215, Université Paris Est Créteil, Université Paris Est, Créteil, France
| | - Paolo Contessotto
- CÚRAM SFI Research Centre for Medical Devices, National University of Ireland Galway, Galway H92 W2TY, Ireland
| | - Dulce Papy-Garcia
- Cell Growth, Tissue Repair and Regeneration (CRRET), UPEC EA 4397/ERL CNRS 9215, Université Paris Est Créteil, Université Paris Est, Créteil, France
| | | | - Michelle Kilcoyne
- CÚRAM SFI Research Centre for Medical Devices, National University of Ireland Galway, Galway H92 W2TY, Ireland; Carbohydrate Signalling Group, Microbiology, School of Natural Sciences, National University of Ireland Galway, Galway H92 W2TY, Ireland
| | - K Schenke-Layland
- Department of Women's Health, Research Institute for Women's Health, The Eberhard-Karls-University Tuebingen, Germany; The Natural and Medical Sciences Institute (NMI) at the University of Tübingen, Reutlingen, Germany
| | - N G Karlsson
- Department of Medical Biochemistry and Cell Biology at Institute of Biomedicine, Sahlgrenska Academy, The University of Gothenburg, Sweden
| | - Karl J A McCullagh
- Physiology Department, National University of Ireland Galway, Galway H92 W2TY, Ireland
| | - Abhay Pandit
- CÚRAM SFI Research Centre for Medical Devices, National University of Ireland Galway, Galway H92 W2TY, Ireland.
| |
Collapse
|
28
|
Prikryl P, Satrapova V, Frydlova J, Hruskova Z, Zima T, Tesar V, Vokurka M. Mass spectrometry-based proteomic exploration of the small urinary extracellular vesicles in ANCA-associated vasculitis in comparison with total urine. J Proteomics 2020; 233:104067. [PMID: 33307252 DOI: 10.1016/j.jprot.2020.104067] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Revised: 09/11/2020] [Accepted: 11/29/2020] [Indexed: 01/07/2023]
Abstract
ANCA-associated vasculitis (AAV) is a rare, but potentially severe autoimmune disease, even nowadays displaying increased mortality and morbidity. Finding early biomarkers of activity and prognosis is thus very important. Small extracellular vesicles (EVs) isolated from urine can be considered as a non-invasive source of biomarkers. We evaluated several protocols for urinary EV isolation. To eliminate contaminating non-vesicular proteins due to AAV associated proteinuria we used proteinase K treatment. We investigated the differences in proteomes of small EVs of patients with AAV compared to healthy controls by label-free LC-MS/MS. In parallel, we performed an analogous proteomic analysis of urine samples from identical patients. The study results showed significant differences and similarities in both EV and urine proteome, the latter one being highly affected by proteinuria. Using bioinformatics tools we explored differentially changed proteins and their related pathways with a focus on the pathophysiology of AAV. Our findings indicate significant regulation of Golgi enzymes, such as MAN1A1, which can be involved in T cell activation by N-glycans glycosylation and may thus play a key role in pathogenesis and diagnosis of AAV. SIGNIFICANCE: The present study explores for the first time the changes in proteomes of small extracellular vesicles and urine of patients with renal ANCA-associated vasculitis compared to healthy controls by label-free LC-MS/MS. Isolation of vesicles from proteinuric urine samples has been modified to minimize contamination by plasma proteins and to reduce co-isolation of extraluminal proteins. Differentially changed proteins and their related pathways with a role in the pathophysiology of AAV were described and discussed. The results could be helpful for the research of potential biomarkers in renal vasculitis associated with ANCA.
Collapse
Affiliation(s)
- Petr Prikryl
- Institute of Pathological Physiology, First Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Veronika Satrapova
- Department of Nephrology, First Faculty of Medicine, Charles University and General University Hospital, Prague, Czech Republic
| | - Jana Frydlova
- Institute of Pathological Physiology, First Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Zdenka Hruskova
- Department of Nephrology, First Faculty of Medicine, Charles University and General University Hospital, Prague, Czech Republic
| | - Tomas Zima
- Institute of Clinical Biochemistry and Laboratory Diagnostics, First Faculty of Medicine, Charles University and General University Hospital, Prague, Czech Republic
| | - Vladimir Tesar
- Department of Nephrology, First Faculty of Medicine, Charles University and General University Hospital, Prague, Czech Republic
| | - Martin Vokurka
- Institute of Pathological Physiology, First Faculty of Medicine, Charles University, Prague, Czech Republic.
| |
Collapse
|
29
|
So J, Ningappa M, Glessner J, Min J, Ashokkumar C, Ranganathan S, Higgs BW, Li D, Sun Q, Schmitt L, Biery AC, Dobrowolski S, Trautz C, Fuhrman L, Schwartz MC, Klena NT, Fusco J, Prasadan K, Adenuga M, Mohamed N, Yan Q, Chen W, Horne W, Dhawan A, Sharif K, Kelly D, Squires RH, Gittes GK, Hakonarson H, Morell V, Lo C, Subramaniam S, Shin D, Sindhi R. Biliary-Atresia-Associated Mannosidase-1-Alpha-2 Gene Regulates Biliary and Ciliary Morphogenesis and Laterality. Front Physiol 2020; 11:538701. [PMID: 33192543 PMCID: PMC7662016 DOI: 10.3389/fphys.2020.538701] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Accepted: 09/23/2020] [Indexed: 01/10/2023] Open
Abstract
Background/Aims Infectious and genetic factors are invoked, respectively in isolated biliary atresia (BA), or syndromic BA, with major extrahepatic anomalies. However, isolated BA is also associated with minor extrahepatic gut and cardiovascular anomalies and multiple susceptibility genes, suggesting common origins. Methods We investigated novel susceptibility genes with genome-wide association, targeted sequencing and tissue staining in BA requiring liver transplantation, independent of BA subtype. Candidate gene effects on morphogenesis, developmental pathways, and ciliogenesis, which regulates left-right patterning were investigated with zebrafish knockdown and mouse knockout models, mouse airway cell cultures, and liver transcriptome analysis. Results Single nucleotide polymorphisms in Mannosidase-1-α-2 (MAN1A2) were significantly associated with BA and with other polymorphisms known to affect MAN1A2 expression but were not differentially enriched in either BA subtype. In zebrafish embryos, man1a2 knockdown caused poor biliary network formation, ciliary dysgenesis in Kupffer’s vesicle, cardiac and liver heterotaxy, and dysregulated egfra and other developmental genes. Suboptimal man1a2 knockdown synergized with suboptimal EGFR signaling or suboptimal knockdown of the EGFR pathway gene, adenosine-ribosylation-factor-6, which had minimal effects individually, to reproduce biliary defects but not heterotaxy. In cultured mouse airway epithelium, Man1a2 knockdown arrested ciliary development and motility. Man1a2–/– mice, which experience respiratory failure, also demonstrated portal and bile ductular inflammation. Human BA liver and Man1a2–/– liver exhibited reduced Man1a2 expression and dysregulated ciliary genes, known to cause multisystem human laterality defects. Conclusion BA requiring transplantation associates with sequence variants in MAN1A2. man1a2 regulates laterality, in addition to hepatobiliary morphogenesis, by regulating ciliogenesis in zebrafish and mice, providing a novel developmental basis for multisystem defects in BA.
Collapse
Affiliation(s)
- Juhoon So
- Department of Developmental Biology, McGowan Institute of Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA, United States
| | - Mylarappa Ningappa
- Hillman Center for Pediatric Transplantation of the Children's Hospital of Pittsburgh of University of Pittsburgh Medical Center (UPMC), Pittsburgh, PA, United States
| | - Joseph Glessner
- Center for Applied Genomics of the Children's Hospital of Philadelphia, Philadelphia, PA, United States
| | - Jun Min
- Departments of Bioengineering, Cellular and Molecular Medicine, and Computer Science and Engineering, University of California San Diego, La Jolla, CA, United States
| | - Chethan Ashokkumar
- Hillman Center for Pediatric Transplantation of the Children's Hospital of Pittsburgh of University of Pittsburgh Medical Center (UPMC), Pittsburgh, PA, United States
| | - Sarangarajan Ranganathan
- Division of Pediatric Pathology, Department of Pathology, Children's Hospital of Pittsburgh of UPMC, Pittsburgh, PA, United States
| | - Brandon W Higgs
- Hillman Center for Pediatric Transplantation of the Children's Hospital of Pittsburgh of University of Pittsburgh Medical Center (UPMC), Pittsburgh, PA, United States
| | - Dong Li
- Center for Applied Genomics of the Children's Hospital of Philadelphia, Philadelphia, PA, United States
| | - Qing Sun
- Hillman Center for Pediatric Transplantation of the Children's Hospital of Pittsburgh of University of Pittsburgh Medical Center (UPMC), Pittsburgh, PA, United States
| | - Lori Schmitt
- Histology Core Laboratory, Children's Hospital of Pittsburgh of UPMC, Pittsburgh, PA, United States
| | - Amy C Biery
- Division of Pediatric Pathology, Department of Pathology, Children's Hospital of Pittsburgh of UPMC, Pittsburgh, PA, United States
| | - Steven Dobrowolski
- Division of Pediatric Pathology, Department of Pathology, Children's Hospital of Pittsburgh of UPMC, Pittsburgh, PA, United States
| | - Christine Trautz
- Hillman Center for Pediatric Transplantation of the Children's Hospital of Pittsburgh of University of Pittsburgh Medical Center (UPMC), Pittsburgh, PA, United States
| | - Leah Fuhrman
- Hillman Center for Pediatric Transplantation of the Children's Hospital of Pittsburgh of University of Pittsburgh Medical Center (UPMC), Pittsburgh, PA, United States
| | | | - Nikolai Thomas Klena
- Department of Developmental Biology, University of Pittsburgh, Pittsburgh, PA, United States
| | - Joseph Fusco
- Pediatric General and Thoracic Surgery, Children's Hospital of Pittsburgh of UPMC, Pittsburgh, PA, United States
| | - Krishna Prasadan
- Pediatric General and Thoracic Surgery, Children's Hospital of Pittsburgh of UPMC, Pittsburgh, PA, United States
| | - Morayooluwa Adenuga
- Hillman Center for Pediatric Transplantation of the Children's Hospital of Pittsburgh of University of Pittsburgh Medical Center (UPMC), Pittsburgh, PA, United States
| | - Nada Mohamed
- Pediatric General and Thoracic Surgery, Children's Hospital of Pittsburgh of UPMC, Pittsburgh, PA, United States
| | - Qi Yan
- Departments of Human Genetics and Biostatistics, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, PA, United States
| | - Wei Chen
- Departments of Human Genetics and Biostatistics, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, PA, United States
| | - William Horne
- Richard King Mellon Foundation Institute for Pediatric Research, Children's Hospital of Pittsburgh of UPMC, Pittsburgh, PA, United States
| | - Anil Dhawan
- Paediatric Liver, GI, and Nutrition, King's College Hospital, London, United Kingdom
| | - Khalid Sharif
- Children's Hospital of Birmingham, Birmingham, United Kingdom
| | - Deirdre Kelly
- Children's Hospital of Birmingham, Birmingham, United Kingdom
| | - Robert H Squires
- Pediatric Gastroenterology, Children's Hospital of Pittsburgh of UPMC, Pittsburgh, PA, United States
| | - George K Gittes
- Pediatric General and Thoracic Surgery, Children's Hospital of Pittsburgh of UPMC, Pittsburgh, PA, United States
| | - Hakon Hakonarson
- Center for Applied Genomics of the Children's Hospital of Philadelphia, Philadelphia, PA, United States
| | - Victor Morell
- Division of Cardiac Surgery, Department of Cardiothoracic Surgery, Children's Hospital of Pittsburgh of UPMC, Pittsburgh, PA, United States
| | - Cecilia Lo
- Department of Developmental Biology, University of Pittsburgh, Pittsburgh, PA, United States
| | - Shankar Subramaniam
- Departments of Bioengineering, Cellular and Molecular Medicine, and Computer Science and Engineering, University of California San Diego, La Jolla, CA, United States
| | - Donghun Shin
- Department of Developmental Biology, McGowan Institute of Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA, United States
| | - Rakesh Sindhi
- Hillman Center for Pediatric Transplantation of the Children's Hospital of Pittsburgh of University of Pittsburgh Medical Center (UPMC), Pittsburgh, PA, United States
| |
Collapse
|
30
|
Spampinato SF, Merlo S, Fagone E, Fruciano M, Sano Y, Kanda T, Sortino MA. Reciprocal Interplay Between Astrocytes and CD4+ Cells Affects Blood-Brain Barrier and Neuronal Function in Response to β Amyloid. Front Mol Neurosci 2020; 13:120. [PMID: 32719583 PMCID: PMC7347984 DOI: 10.3389/fnmol.2020.00120] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Accepted: 06/10/2020] [Indexed: 12/14/2022] Open
Abstract
Background: In Alzheimer’s disease (AD) neuronal degeneration is associated with gliosis and infiltration of peripheral blood mononuclear cells (PBMCs), which participate in neuroinflammation. Defects at the blood-brain barrier (BBB) facilitate PBMCs migration towards the central nervous system (CNS) and in particular CD4+ T cells have been found in areas severely affected in AD. However, the role of T cells, once they migrate into the CNS, is not well defined. CD4+ cells interact with astrocytes able to release several factors and cytokines that can modulate T cell polarization; similarly, astrocytic properties are modulated after interaction with T cells. Methods: In in vitro models, astrocytes were primed with β-amyloid (Aβ; 2.5 μM, 5 h) and then co-cultured with magnetically isolated CD4+ cells. Cytokines expression was evaluated both in co-cultured CD4+ cells and astrocytes. The effects of this crosstalk were further evaluated by co-culturing CD4+ cells with the neuronal-like SH-SY5Y cell line and astrocytes with endothelial cells. Results: The pattern of cytokines and trophic factors expressed by CD4+ cells were strongly modulated in the presence of Aβ-primed astrocytes. Specifically, the percentage of IL-4+ and IFNγ+ CD4+ cells was significantly increased and reduced, respectively. Further, increased BDNF mRNA levels were observed in CD4+ cells. When SH-SY5Y cells were co-cultured with astrocyte-conditioned CD4+ cells and exposed to Aβ, the reduction of the presynaptic protein synaptophysin was prevented with a BDNF-dependent mechanism. In astrocytes co-cultured with CD4+ cells, reduced mRNA levels of inflammatory cytokines and VEGF were observed. This was paralleled by the prevention of the reduction of claudin-5 when astrocytes were co-cultured with endothelial cells. Conclusion: Following Aβ exposure, there exists reciprocal crosstalk between infiltrating peripheral cells and astrocytes that in turn affects not only endothelial function and thus BBB properties, but also neuronal behavior. Since astrocytes are the first cells that lymphocytes interact with and are among the principal players in neuroinflammation occurring in AD, understanding this crosstalk may disclose new potential targets of intervention in the treatment of neurodegeneration.
Collapse
Affiliation(s)
- Simona Federica Spampinato
- Section of Pharmacology, Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
| | - Sara Merlo
- Section of Pharmacology, Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
| | - Evelina Fagone
- Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy
| | - Mary Fruciano
- Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy
| | - Yasuteru Sano
- Department of Neurology and Clinical Neuroscience, Yamaguchi University Graduate School of Medicine, Ube, Japan
| | - Takashi Kanda
- Department of Neurology and Clinical Neuroscience, Yamaguchi University Graduate School of Medicine, Ube, Japan
| | - Maria Angela Sortino
- Section of Pharmacology, Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
| |
Collapse
|
31
|
Hydrogen peroxide regulates endothelial surface N-glycoforms to control inflammatory monocyte rolling and adhesion. Redox Biol 2020; 34:101498. [PMID: 32171755 PMCID: PMC7327964 DOI: 10.1016/j.redox.2020.101498] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2020] [Revised: 03/02/2020] [Accepted: 03/04/2020] [Indexed: 12/15/2022] Open
Abstract
Monocyte extravasation through the endothelial layer is a hallmark of atherosclerotic plaque development and is mediated by heavily N-glycosylated surface adhesion molecules, such as intercellular adhesion molecule-1 (ICAM-1). N-glycosylation is a key co- and post-translational modification that adds sugar molecules to Asparagine residues of surface and secreted proteins. While it has been suggested that surface and secreted proteins will not be expressed unless fully processed to a complex N-glycoform, emerging data has suggested that multiple N-glycoforms can exist on the cell surface. Previous data from our lab has shown that endothelial inflammation produces multiple N-glycoforms of ICAM-1, and that a hypoglycosylated, or high-mannose (HM), form of ICAM-1 enhances adhesion of pro-inflammatory monocytes associated with more severe atherosclerosis and adverse cardiac events. Despite these findings, little is understood about the regulation of N-glycans during disease. In this study, we focus on the α-mannosidases; an understudied class of enzymes for early N-glycan processing. We show that α-mannosidase activity decreases with TNFα treatment in endothelial cells, and this decrease correlates with HM N-glycan formation on the cell surface. Further, we demonstrate that this inhibition is class-I dependent, and is independent of NF-κB upregulation of ICAM-1. Finally, we show that this inhibition is due in part to hydrogen peroxide (H2O2), generated by Endoplasmic Reticulum oxidoreductase 1-α (ERO1α). These data provide insights into the regulation of surface N-glycans during inflammation and demonstrate a novel role for reactive species in N-glycan biosynthesis.
Collapse
|
32
|
Regal-McDonald K, Patel RP. Selective Recruitment of Monocyte Subsets by Endothelial N-Glycans. THE AMERICAN JOURNAL OF PATHOLOGY 2020; 190:947-957. [PMID: 32084367 DOI: 10.1016/j.ajpath.2020.01.006] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/01/2019] [Revised: 12/17/2019] [Accepted: 01/07/2020] [Indexed: 12/26/2022]
Abstract
Monocyte rolling, adhesion, and transmigration across the endothelium are mediated by specific interactions between surface adhesion molecules. This process is fundamental to innate immunity and to inflammatory disease, including atherosclerosis, where monocyte egress into the intimal space is central to formation of fatty plaques. Monocytes are a heterogeneous population of three distinct subsets of cells, all of which play different roles in atherosclerosis progression. However, it is not well understood how interactions between different monocyte subsets and the endothelium are regulated. Furthermore, it is appreciated that endothelial adhesion molecules are heavily N-glycosylated, but beyond regulating protein trafficking to the cell surface, whether and if so how these N-glycans contribute to monocyte recruitment is not known. This review discusses how changes in endothelial N-glycosylation may impact vascular and monocytic inflammation. It will also discuss how regulating N-glycoforms on the endothelial surface may allow for the recruitment of specific monocyte subsets to sites of inflammation, and how further understanding in this area may lead to the development of glyco-specific therapeutics in the treatment of cardiovascular disease.
Collapse
Affiliation(s)
- Kellie Regal-McDonald
- Department of Pathology, University of Alabama at Birmingham, Birmingham, Alabama; Center for Free Radical Biology, University of Alabama at Birmingham, Birmingham, Alabama
| | - Rakesh P Patel
- Department of Pathology, University of Alabama at Birmingham, Birmingham, Alabama; Center for Free Radical Biology, University of Alabama at Birmingham, Birmingham, Alabama.
| |
Collapse
|
33
|
Regal-McDonald K, Xu B, Barnes JW, Patel RP. High-mannose intercellular adhesion molecule-1 enhances CD16 + monocyte adhesion to the endothelium. Am J Physiol Heart Circ Physiol 2019; 317:H1028-H1038. [PMID: 31398058 DOI: 10.1152/ajpheart.00306.2019] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Human monocytes have been classified into three distinct groups, classical (anti-inflammatory; CD14+/CD16-), nonclassical (patrolling; CD14+/CD16++), and intermediate (proinflammatory; CD14++/CD16+). Adhesion of nonclassical/intermediate monocytes with the endothelium is important for innate immunity, and also vascular inflammatory disease. However, there is an incomplete understanding of the mechanisms that regulate CD16+ versus CD16- monocyte adhesion to the inflamed endothelium. Here, we tested the hypothesis that a high-mannose (HM) N-glycoform of intercellular adhesion molecule-1 (ICAM-1) on the endothelium mediates the selective recruitment of CD16+ monocytes. Using TNF-α treatment of human umbilical vein endothelial cells (HUVECs), and using proximity ligation assay for detecting proximity of specific N-glycans and ICAM-1, we show that TNF-α induces HM-ICAM-1 formation on the endothelial surface in a time-dependent manner. We next measured CD16- or CD16+ monocyte rolling and adhesion to TNF-α-treated HUVECs in which HM- or hybrid ICAM-1 N-glycoforms were generated using the α-mannosidase class I and II inhibitors, kifunensine and swainsonine, respectively. Expression of HM-ICAM-1 selectively enhanced CD16+ monocyte adhesion under flow with no effect on CD16- monocytes noted. CD16+ monocyte adhesion was abrogated by blocking either HM epitopes or ICAM-1. A critical role for HM-ICAM-1 in mediating CD16+ monocyte rolling and adhesion was confirmed using COS-1 cells engineered to express HM or complex ICAM-1 N-glycoforms. These data suggest that HM-ICAM-1 selectively recruits nonclassical/intermediate CD16+ monocytes to the activated endothelium.NEW & NOTEWORTHY Monocyte subsets have been associated with cardiovascular disease, yet it is unknown how different subsets are recruited to the endothelium. This study demonstrates the formation of distinct ICAM-1 N-glycoforms in the activated endothelium and reveals a key role for high mannose ICAM-1 in mediating proinflammatory CD16+ monocyte adhesion. Presented data identify roles for endothelial N-glycans in recruiting specific monocyte subsets during inflammation.
Collapse
Affiliation(s)
- Kellie Regal-McDonald
- Department of Pathology, University of Alabama at Birmingham, Birmingham, Alabama.,Center for Free Radical Biology, University of Alabama at Birmingham, Birmingham, Alabama
| | - Brittney Xu
- Department of Pathology, University of Alabama at Birmingham, Birmingham, Alabama
| | - Jarrod W Barnes
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama
| | - Rakesh P Patel
- Department of Pathology, University of Alabama at Birmingham, Birmingham, Alabama.,Center for Free Radical Biology, University of Alabama at Birmingham, Birmingham, Alabama
| |
Collapse
|
34
|
Spampinato SF, Merlo S, Fagone E, Fruciano M, Barbagallo C, Kanda T, Sano Y, Purrello M, Vancheri C, Ragusa M, Sortino MA. Astrocytes Modify Migration of PBMCs Induced by β-Amyloid in a Blood-Brain Barrier in vitro Model. Front Cell Neurosci 2019; 13:337. [PMID: 31396056 PMCID: PMC6664149 DOI: 10.3389/fncel.2019.00337] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2019] [Accepted: 07/10/2019] [Indexed: 12/20/2022] Open
Abstract
Background The brain is protected by the blood-brain barrier (BBB), constituted by endothelial cells supported by pericytes and astrocytes. In Alzheimer’s disease a dysregulation of the BBB occurs since the early phases of the disease leading to an increased access of solutes and immune cells that can participate to the central inflammatory response. Here we investigated whether astrocytes may influence endothelial-leukocytes interaction in the presence of amyloid-β (Aβ). Methods We used an in vitro BBB model, where endothelial cells, cultured alone or with astrocytes were exposed for 5 h to Aβ, both under resting or inflammatory conditions (TNFα and IFNγ), to evaluate endothelial barrier properties, as well as transendothelial migration of peripheral blood mononuclear cells (PBMCs). Results In the co-culture model, barrier permeability to solutes was increased by all treatments, but migration was only observed in inflammatory conditions and was prevented by Aβ treatment. On the contrary, in endothelial monocultures, Aβ induced leukocytes migration under resting conditions and did not modify that induced by inflammatory cytokines. In endothelial astrocyte co-cultures, a low molecular weight (MW) isoform of the adhesion molecule ICAM-1, important to allow interaction with PBMCs, was increased after 5 h exposure to inflammatory cytokines, an effect that was prevented by Aβ. This modulation by Aβ was not observed in endothelial monocultures. In addition, endothelial expression of β-1,4-N-acetylglucosaminyltransferase III (Gnt-III), responsible for the formation of the low MW ICAM-1 isoform, was enhanced in inflammatory conditions, but negatively modulated by Aβ only in the co-culture model. miR-200b, increased in astrocytes following Aβ treatment and may represent one of the factors involved in the control of Gnt-III expression. Conclusion These data point out that, at least in the early phases of Aβ exposure, astrocytes play a role in the modulation of leukocytes migration through the endothelial layer.
Collapse
Affiliation(s)
- Simona Federica Spampinato
- Section of Pharmacology, Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
| | - Sara Merlo
- Section of Pharmacology, Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
| | - Evelina Fagone
- Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy
| | - Mary Fruciano
- Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy
| | - Cristina Barbagallo
- Section of Biology and Genetics, Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
| | - Takashi Kanda
- Department of Neurology and Clinical Neuroscience, Yamaguchi University, Yamaguchi, Japan
| | - Yasuteru Sano
- Department of Neurology and Clinical Neuroscience, Yamaguchi University, Yamaguchi, Japan
| | - Michele Purrello
- Section of Biology and Genetics, Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
| | - Carlo Vancheri
- Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy
| | - Marco Ragusa
- Section of Biology and Genetics, Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy.,Oasi Research Institute - IRCCS, Troina, Italy
| | - Maria Angela Sortino
- Section of Pharmacology, Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
| |
Collapse
|
35
|
Chandler KB, Costello CE, Rahimi N. Glycosylation in the Tumor Microenvironment: Implications for Tumor Angiogenesis and Metastasis. Cells 2019; 8:E544. [PMID: 31195728 PMCID: PMC6627046 DOI: 10.3390/cells8060544] [Citation(s) in RCA: 71] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2019] [Revised: 05/31/2019] [Accepted: 06/01/2019] [Indexed: 01/27/2023] Open
Abstract
Just as oncogene activation and tumor suppressor loss are hallmarks of tumor development, emerging evidence indicates that tumor microenvironment-mediated changes in glycosylation play a crucial functional role in tumor progression and metastasis. Hypoxia and inflammatory events regulate protein glycosylation in tumor cells and associated stromal cells in the tumor microenvironment, which facilitates tumor progression and also modulates a patient's response to anti-cancer therapeutics. In this review, we highlight the impact of altered glycosylation on angiogenic signaling and endothelial cell adhesion, and the critical consequences of these changes in tumor behavior.
Collapse
Affiliation(s)
- Kevin Brown Chandler
- Center for Biomedical Mass Spectrometry, Department of Biochemistry, Boston University School of Medicine, Boston, MA 02118, USA.
| | - Catherine E Costello
- Center for Biomedical Mass Spectrometry, Department of Biochemistry, Boston University School of Medicine, Boston, MA 02118, USA.
| | - Nader Rahimi
- Department of Pathology and Laboratory Medicine, Boston University School of Medicine, Boston, MA 02118, USA.
| |
Collapse
|
36
|
Abstract
The glycome describes the complete repertoire of glycoconjugates composed of carbohydrate chains, or glycans, that are covalently linked to lipid or protein molecules. Glycoconjugates are formed through a process called glycosylation and can differ in their glycan sequences, the connections between them and their length. Glycoconjugate synthesis is a dynamic process that depends on the local milieu of enzymes, sugar precursors and organelle structures as well as the cell types involved and cellular signals. Studies of rare genetic disorders that affect glycosylation first highlighted the biological importance of the glycome, and technological advances have improved our understanding of its heterogeneity and complexity. Researchers can now routinely assess how the secreted and cell-surface glycomes reflect overall cellular status in health and disease. In fact, changes in glycosylation can modulate inflammatory responses, enable viral immune escape, promote cancer cell metastasis or regulate apoptosis; the composition of the glycome also affects kidney function in health and disease. New insights into the structure and function of the glycome can now be applied to therapy development and could improve our ability to fine-tune immunological responses and inflammation, optimize the performance of therapeutic antibodies and boost immune responses to cancer. These examples illustrate the potential of the emerging field of 'glycomedicine'.
Collapse
Affiliation(s)
- Colin Reily
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Tyler J Stewart
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, AL, USA
- Department of Biochemistry and Molecular Genetics, University of Alabama at Birmingham, Birmingham, AL, USA
- National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD, USA
| | - Matthew B Renfrow
- Department of Biochemistry and Molecular Genetics, University of Alabama at Birmingham, Birmingham, AL, USA.
| | - Jan Novak
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, AL, USA.
| |
Collapse
|
37
|
Liu X, Li A, Ju Y, Liu W, Shi H, Hu R, Zhou Z, Sun X. β4GalT1 Mediates PPARγ N-Glycosylation to Attenuate Microglia Inflammatory Activation. Inflammation 2018; 41:1424-1436. [DOI: 10.1007/s10753-018-0789-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
|
38
|
Protein N-Glycosylation in Cardiovascular Diseases and Related Risk Factors. CURRENT CARDIOVASCULAR RISK REPORTS 2018. [DOI: 10.1007/s12170-018-0579-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
|
39
|
Guipaud O, Jaillet C, Clément-Colmou K, François A, Supiot S, Milliat F. The importance of the vascular endothelial barrier in the immune-inflammatory response induced by radiotherapy. Br J Radiol 2018; 91:20170762. [PMID: 29630386 DOI: 10.1259/bjr.20170762] [Citation(s) in RCA: 64] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Altered by ionising radiation, the vascular network is considered as a prime target to limit normal tissue damage and improve tumour control in radiotherapy (RT). Irradiation damages and/or activates endothelial cells, which then participate in the recruitment of circulating cells, especially by overexpressing cell adhesion molecules, but also by other as yet unknown mechanisms. Radiation-induced lesions are associated with infiltration of immune-inflammatory cells from the blood and/or the lymph circulation. Damaged cells from the tissues and immune-inflammatory resident cells release factors that attract cells from the circulation, leading to the restoration of tissue balance by fighting against infection, elimination of damaged cells and healing of the injured area. In normal tissues that surround the tumours, the development of an immune-inflammatory reaction in response to radiation-induced tissue injury can turn out to be chronic and deleterious for the organ concerned, potentially leading to fibrosis and/or necrosis of the irradiated area. Similarly, tumours can elicit an immune-inflammation reaction, which can be initialised and amplified by cancer therapy such as radiotherapy, although immune checkpoints often allow many cancers to be protected by inhibiting the T-cell signal. Herein, we have explored the involvement of vascular endothelium in the fate of healthy tissues and tumours undergoing radiotherapy. This review also covers current investigations that take advantage of the radiation-induced response of the vasculature to spare healthy tissue and/or target tumours better.
Collapse
Affiliation(s)
- Olivier Guipaud
- 1 Human Health Department, Institut de Radioprotection et de Sûreté Nucléaire (IRSN), PSE-SANTE, SERAMED, LRMed , Fontenay-aux-Roses , France
| | - Cyprien Jaillet
- 1 Human Health Department, Institut de Radioprotection et de Sûreté Nucléaire (IRSN), PSE-SANTE, SERAMED, LRMed , Fontenay-aux-Roses , France
| | - Karen Clément-Colmou
- 2 Département de Radiothérapie, Institut de Cancérologie de l'Ouest , Nantes St-Herblain , France.,3 Oncology and New Concept in Oncology Department, Centre de Recherche en Cancérologie et Immunologie Nantes-Angers (CRCiNA), Unité U1232, Institut de Recherche en Santé de l'Université de Nantes , Nantes , France
| | - Agnès François
- 1 Human Health Department, Institut de Radioprotection et de Sûreté Nucléaire (IRSN), PSE-SANTE, SERAMED, LRMed , Fontenay-aux-Roses , France
| | - Stéphane Supiot
- 2 Département de Radiothérapie, Institut de Cancérologie de l'Ouest , Nantes St-Herblain , France.,3 Oncology and New Concept in Oncology Department, Centre de Recherche en Cancérologie et Immunologie Nantes-Angers (CRCiNA), Unité U1232, Institut de Recherche en Santé de l'Université de Nantes , Nantes , France
| | - Fabien Milliat
- 1 Human Health Department, Institut de Radioprotection et de Sûreté Nucléaire (IRSN), PSE-SANTE, SERAMED, LRMed , Fontenay-aux-Roses , France
| |
Collapse
|
40
|
Reduced mannosidase MAN1A1 expression leads to aberrant N-glycosylation and impaired survival in breast cancer. Br J Cancer 2018; 118:847-856. [PMID: 29381688 PMCID: PMC5877434 DOI: 10.1038/bjc.2017.472] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2017] [Revised: 11/28/2017] [Accepted: 11/29/2017] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Alterations in protein glycosylation have been related to malignant transformation and tumour progression. We recently showed that low mRNA levels of Golgi alpha-mannosidase MAN1A1 correlate with poor prognosis in breast cancer patients. METHODS We analysed the role of MAN1A1 on a protein level using western blot analysis (n=105) and studied the impact of MAN1A1-related glycosylation on the prognostic relevance of adhesion molecules involved in breast cancer using microarray data (n=194). Functional consequences of mannosidase inhibition using the inhibitor kifunensine or MAN1A1 silencing were investigated in breast cancer cells in vitro. RESULTS Patients with low/moderate MAN1A1 expression in tumours showed significantly shorter disease-free intervals than those with high MAN1A1 levels (P=0.005). Moreover, low MAN1A1 expression correlated significantly with nodal status, grading and brain metastasis. At an mRNA level, membrane proteins ALCAM and CD24 were only significantly prognostic in tumours with high MAN1A1 expression. In vitro, reduced MAN1A1 expression or mannosidase inhibition led to a significantly increased adhesion of breast cancer cells to endothelial cells. CONCLUSIONS Our study demonstrates the prognostic role of MAN1A1 in breast cancer by affecting the adhesive properties of tumour cells and the strong influence of this glycosylation enzyme on the prognostic impact of some adhesion proteins.
Collapse
|
41
|
Jaillet C, Morelle W, Slomianny MC, Paget V, Tarlet G, Buard V, Selbonne S, Caffin F, Rannou E, Martinez P, François A, Foulquier F, Allain F, Milliat F, Guipaud O. Radiation-induced changes in the glycome of endothelial cells with functional consequences. Sci Rep 2017; 7:5290. [PMID: 28706280 PMCID: PMC5509684 DOI: 10.1038/s41598-017-05563-y] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2017] [Accepted: 05/30/2017] [Indexed: 12/27/2022] Open
Abstract
As it is altered by ionizing radiation, the vascular network is considered as a prime target in limiting normal tissue damage and improving tumor control in radiation therapy. Irradiation activates endothelial cells which then participate in the recruitment of circulating cells, especially by overexpressing cell adhesion molecules, but also by other as yet unknown mechanisms. Since protein glycosylation is an important determinant of cell adhesion, we hypothesized that radiation could alter the glycosylation pattern of endothelial cells and thereby impact adhesion of circulating cells. Herein, we show that ionizing radiation increases high mannose-type N-glycans and decreases glycosaminoglycans. These changes stimulate interactions measured under flow conditions between irradiated endothelial cells and monocytes. Targeted transcriptomic approaches in vitro in endothelial cells and in vivo in a radiation enteropathy mouse model confirm that genes involved in N- and O-glycosylation are modulated by radiation, and in silico analyses give insight into the mechanism by which radiation modifies glycosylation. The endothelium glycome may therefore be considered as a key therapeutic target for modulating the chronic inflammatory response observed in healthy tissues or for participating in tumor control by radiation therapy.
Collapse
Affiliation(s)
- Cyprien Jaillet
- Institute for Radiological Protection and Nuclear Safety (IRSN), PRP-HOM, SRBE, L3R, 92260, Fontenay-aux-Roses, France
| | - Willy Morelle
- University of Lille, CNRS, UMR 8576 - UGSF - Unité de Glycobiologie Structurale et Fonctionnelle, 59000, Lille, France
| | - Marie-Christine Slomianny
- University of Lille, CNRS, UMR 8576 - UGSF - Unité de Glycobiologie Structurale et Fonctionnelle, 59000, Lille, France
| | - Vincent Paget
- Institute for Radiological Protection and Nuclear Safety (IRSN), PRP-HOM, SRBE, L3R, 92260, Fontenay-aux-Roses, France
| | - Georges Tarlet
- Institute for Radiological Protection and Nuclear Safety (IRSN), PRP-HOM, SRBE, L3R, 92260, Fontenay-aux-Roses, France
| | - Valérie Buard
- Institute for Radiological Protection and Nuclear Safety (IRSN), PRP-HOM, SRBE, L3R, 92260, Fontenay-aux-Roses, France
| | - Sonia Selbonne
- Institute for Radiological Protection and Nuclear Safety (IRSN), PRP-HOM, SRBE, L3R, 92260, Fontenay-aux-Roses, France
| | - Fanny Caffin
- Institute for Radiological Protection and Nuclear Safety (IRSN), PRP-HOM, SRBE, L3R, 92260, Fontenay-aux-Roses, France
| | - Emilie Rannou
- Institute for Radiological Protection and Nuclear Safety (IRSN), PRP-HOM, SRBE, L3R, 92260, Fontenay-aux-Roses, France.,Department of Molecular, Cell and Developmental Biology, UCLA, CA 90095-7239, Los Angeles, USA
| | - Pierre Martinez
- University of Lille, CNRS, UMR 8576 - UGSF - Unité de Glycobiologie Structurale et Fonctionnelle, 59000, Lille, France.,GSK - GlaxoSmithKline, 1300, Wavre, Belgium
| | - Agnès François
- Institute for Radiological Protection and Nuclear Safety (IRSN), PRP-HOM, SRBE, L3R, 92260, Fontenay-aux-Roses, France
| | - François Foulquier
- University of Lille, CNRS, UMR 8576 - UGSF - Unité de Glycobiologie Structurale et Fonctionnelle, 59000, Lille, France
| | - Fabrice Allain
- University of Lille, CNRS, UMR 8576 - UGSF - Unité de Glycobiologie Structurale et Fonctionnelle, 59000, Lille, France
| | - Fabien Milliat
- Institute for Radiological Protection and Nuclear Safety (IRSN), PRP-HOM, SRBE, L3R, 92260, Fontenay-aux-Roses, France
| | - Olivier Guipaud
- Institute for Radiological Protection and Nuclear Safety (IRSN), PRP-HOM, SRBE, L3R, 92260, Fontenay-aux-Roses, France.
| |
Collapse
|
42
|
Barel M, Charbit A. Role of Glycosylation/Deglycolysation Processes in Francisella tularensis Pathogenesis. Front Cell Infect Microbiol 2017; 7:71. [PMID: 28377902 PMCID: PMC5359314 DOI: 10.3389/fcimb.2017.00071] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2017] [Accepted: 02/27/2017] [Indexed: 12/15/2022] Open
Abstract
Francisella tularensis is able to invade, survive and replicate inside a variety of cell types. However, in vivo F. tularensis preferentially enters host macrophages where it rapidly escapes to the cytosol to avoid phagosomal stresses and to multiply to high numbers. We previously showed that human monocyte infection by F. tularensis LVS triggered deglycosylation of the glutamine transporter SLC1A5. However, this deglycosylation, specifically induced by Francisella infection, was not restricted to SLC1A5, suggesting that host protein deglycosylation processes in general might contribute to intracellular bacterial adaptation. Indeed, we later found that Francisella infection modulated the transcription of numerous glycosidase and glycosyltransferase genes in human macrophages and analysis of cell extracts revealed an important increase of N and O-protein glycosylation. In eukaryotic cells, glycosylation has significant effects on protein folding, conformation, distribution, stability, and activity and dysfunction of protein glycosylation may lead to development of diseases like cancer and pathogenesis of infectious diseases. Pathogenic bacteria have also evolved dedicated glycosylation machineries and have notably been shown to use these glycoconjugates as ligands to specifically interact with the host. In this review, we will focus on Francisella and summarize our current understanding of the importance of these post-translational modifications on its intracellular niche adaptation.
Collapse
Affiliation(s)
- Monique Barel
- Sorbonne Paris Cité, Bâtiment Leriche, Université Paris DescartesParis, France; Institut National de la Santé et de la Recherche Médicale, Institut Necker-Enfants Malades, INSERM U1151 -Team 11, Pathogenesis of Systemic InfectionsParis, France; Centre National de la Recherche Scientifique, UMR8253Paris, France
| | - Alain Charbit
- Sorbonne Paris Cité, Bâtiment Leriche, Université Paris DescartesParis, France; Institut National de la Santé et de la Recherche Médicale, Institut Necker-Enfants Malades, INSERM U1151 -Team 11, Pathogenesis of Systemic InfectionsParis, France; Centre National de la Recherche Scientifique, UMR8253Paris, France
| |
Collapse
|
43
|
Hu S, Jiang LB, Zou XJ, Yi W, Tian DY. Hepatitis B virus upregulates host expression of α-1,2-mannosidases via the PPARα pathway. World J Gastroenterol 2016; 22:9534-9543. [PMID: 27920474 PMCID: PMC5116597 DOI: 10.3748/wjg.v22.i43.9534] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/24/2016] [Revised: 09/20/2016] [Accepted: 10/19/2016] [Indexed: 02/06/2023] Open
Abstract
AIM To assess the effects of hepatitis B virus (HBV) on the expression of host α-1,2-mannosidases and determine the underlying mechanisms.
METHODS We measured the expression levels of MAN1A1, MAN1A2, MAN1B1, and MAN1C1 in cell lines HepG2.2.15, HepN10, HepAD38 and HepG2 by Western blot. Viral antigens (HBsAg and HBeAg) in the culture medium were measured using the chemiluminescence method. HBV DNA quantification assays were performed using a commercial real-time PCR kit. Protein levels of human liver tissue α-1,2-mannosidases were also evaluated by Western blot. Plasmids containing seven individual viral genes of HBV (PTT22-HBx, PTT22-HBs, PTT22-preS2, PTT22-preS1, PTT22-HBc, PTT22-HBe, and PTT22-HBp) or control plasmids (PTT22-vector) were transfected into HepG2 cells. MK886 (PPARα) and GW9662 (PPARγ) inhibitors were used to explore the effects of HBV on α-1,2-mannosidase expression after the PPARα and PPARγ pathways were blocked.
RESULTS We showed that the expression of α-1,2-mannosidases was higher in stably transfected HBV cells than in controls. The expression levels of α-1,2-mannosidase were higher in AD38 cells than those in ND10 cells, which were in turn greater than those in G2.2.15 cells, and positively correlated with the expression of HBsAg in all the cell lines. Levels of α-1,2-mannosidase in non-tumorous liver tissues of HBV-related HCC patients were also higher than in the tissues from non-HBV-related HCC patients. Moreover, transfecting HepG2 cells with a component of the HBV viral envelope also increased the expression of α-1,2-mannosidases. However, this envelope protein component could not induce MAN1C1 expression in the presence of a PPARα inhibitor, MK886. We also found that MK886 did not affect the expression of MAN1C1 in AD38 cells without tetracycline in the culture medium. This phenomenon was not observed in the case of GW9662.
CONCLUSION Our results indicate that HBV increases the expression of α-mannosidases both in vitro and in vivo via activation of the PPARα pathway by its envelope protein.
Collapse
|
44
|
Goltsman I, Khoury EE, Winaver J, Abassi Z. Does Thiazolidinedione therapy exacerbate fluid retention in congestive heart failure? Pharmacol Ther 2016; 168:75-97. [PMID: 27598860 DOI: 10.1016/j.pharmthera.2016.09.007] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The ever-growing global burden of congestive heart failure (CHF) and type 2 diabetes mellitus (T2DM) as well as their co-existence necessitate that anti-diabetic pharmacotherapy will modulate the cardiovascular risk inherent to T2DM while complying with the accompanying restrictions imposed by CHF. The thiazolidinedione (TZD) family of peroxisome proliferator-activated receptor γ (PPARγ) agonists initially provided a promising therapeutic option in T2DM owing to anti-diabetic efficacy combined with pleiotropic beneficial cardiovascular effects. However, the utility of TZDs in T2DM has declined in the past decade, largely due to concomitant adverse effects of fluid retention and edema formation attributed to salt-retaining effects of PPARγ activation on the nephron. Presumably, the latter effects are potentially deleterious in the context of pre-existing fluid retention in CHF. However, despite a considerable body of evidence on mechanisms responsible for TZD-induced fluid retention suggesting that this class of drugs is rightfully prohibited from use in CHF patients, there is a paucity of experimental and clinical studies that investigate the effects of TZDs on salt and water homeostasis in the CHF setting. In an attempt to elucidate whether TZDs actually exacerbate the pre-existing fluid retention in CHF, our review summarizes the pathophysiology of fluid retention in CHF. Moreover, we thoroughly review the available data on TZD-induced fluid retention and proposed mechanisms in animals and patients. Finally, we will present recent studies challenging the common notion that TZDs worsen renal salt and water retention in CHF.
Collapse
Affiliation(s)
- Ilia Goltsman
- Department of Physiology, Biophysics and Systems Biology, The Bruce Rappaport, Rappaport Faculty of Medicine, Technion, Haifa, Israel
| | - Emad E Khoury
- Department of Physiology, Biophysics and Systems Biology, The Bruce Rappaport, Rappaport Faculty of Medicine, Technion, Haifa, Israel
| | - Joseph Winaver
- Department of Physiology, Biophysics and Systems Biology, The Bruce Rappaport, Rappaport Faculty of Medicine, Technion, Haifa, Israel
| | - Zaid Abassi
- Department of Physiology, Biophysics and Systems Biology, The Bruce Rappaport, Rappaport Faculty of Medicine, Technion, Haifa, Israel; Department of Laboratory Medicine, Rambam Human Health Care Campus, Haifa, Israel.
| |
Collapse
|
45
|
Čejková S, Králová Lesná I, Poledne R. Monocyte adhesion to the endothelium is an initial stage of atherosclerosis development. COR ET VASA 2016. [DOI: 10.1016/j.crvasa.2015.08.002] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
|
46
|
Calimlioglu B, Karagoz K, Sevimoglu T, Kilic E, Gov E, Arga KY. Tissue-Specific Molecular Biomarker Signatures of Type 2 Diabetes: An Integrative Analysis of Transcriptomics and Protein-Protein Interaction Data. OMICS-A JOURNAL OF INTEGRATIVE BIOLOGY 2016; 19:563-73. [PMID: 26348713 DOI: 10.1089/omi.2015.0088] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Type 2 diabetes mellitus is a major global public health burden. A complex metabolic disease, type 2 diabetes affects multiple different tissues, demanding a "systems medicine" approach to biomarker and novel diagnostic discovery, not to mention data integration across omics-es. In the present study, transcriptomics data from different tissues including beta-cells, pancreatic islets, arterial tissue, peripheral blood mononuclear cells, liver, and skeletal muscle of 228 samples were integrated with protein-protein interaction data and genome scale metabolic models to unravel the molecular and tissue-specific biomarker signatures of type 2 diabetes mellitus. Classifying differentially expressed genes, reconstruction and topological analysis of active protein-protein interaction subnetworks indicated that genomic reprogramming depends on the type of tissue, whereas there are common signatures at different levels. Among all tissue and cell types, Mannosidase Alpha Class 1A Member 2 was the common signature at genome level, and activation-ppara reaction, which stimulates a nuclear receptor protein, was found out as the mutual reporter at metabolic level. Moreover, miR-335 and miR-16-5p came into prominence in regulation of transcription at different tissues. On the other hand, distinct signatures were observed for different tissues at the metabolome level. Various coenzyme-A derivatives were significantly enriched metabolites in pancreatic islets, whereas skeletal muscle was enriched for cholesterol, malate, L-carnitine, and several amino acids. Results have showed utmost importance concerning relations between T2D and cancer, blood coagulation, neurodegenerative diseases, and specific metabolic and signaling pathways.
Collapse
Affiliation(s)
- Beste Calimlioglu
- 1 Department of Bioengineering, Marmara University , Istanbul, Turkey .,2 Department of Bioengineering, Istanbul Medeniyet University , Istanbul, Turkey
| | - Kubra Karagoz
- 1 Department of Bioengineering, Marmara University , Istanbul, Turkey
| | - Tuba Sevimoglu
- 1 Department of Bioengineering, Marmara University , Istanbul, Turkey
| | - Elif Kilic
- 1 Department of Bioengineering, Marmara University , Istanbul, Turkey
| | - Esra Gov
- 1 Department of Bioengineering, Marmara University , Istanbul, Turkey
| | - Kazim Yalcin Arga
- 1 Department of Bioengineering, Marmara University , Istanbul, Turkey
| |
Collapse
|
47
|
Barel M, Harduin-Lepers A, Portier L, Slomianny MC, Charbit A. Host glycosylation pathways and the unfolded protein response contribute to the infection by Francisella. Cell Microbiol 2016; 18:1763-1781. [PMID: 27185209 DOI: 10.1111/cmi.12614] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2015] [Revised: 05/09/2016] [Accepted: 05/09/2016] [Indexed: 12/14/2022]
Abstract
Protein glycosylation processes play a crucial role in most physiological functions, including cell signalling, cellular differentiation and adhesion. We previously demonstrated that rapid deglycosylation of membrane proteins was specifically triggered after infection of human macrophages by the bacterial pathogen Francisella tularensis. Using a glycan processing gene microarray, we found here that Francisella infection modulated expression of numerous glycosidase and glycosyltransferase genes. Furthermore, analysis of cell extracts from infected macrophages by Lectin and Western blotting revealed an important increase of N- and O-protein glycosylation. We chose to focus in the present work on one of the O-glycosylated proteins identified by mass spectrometry, the multifunctional endoplasmic reticulum chaperone BiP (HSPA5/GRP78). We demonstrate that BiP expression is modulated upon Francisella infection and is required to support its intracellular multiplication. Moreover, we show that Francisella differentially modulates the BiP-dependent activation of three key proteins of the unfolded protein response (UPR), IRE1, PERK and ATF6. The effects exerted on human cells by Francisella may thus constitute a novel excample of UPR manipulation contributing to intracellular bacterial adaptation.
Collapse
Affiliation(s)
- Monique Barel
- Université Paris Descartes, Sorbonne Paris Cité, Paris, France.,Institut Necker Enfants-Malades INSERM, U1151, Team 11, Unité de Pathogénie des Infections Systémiques, Paris, France
| | - Anne Harduin-Lepers
- Univ. Lille, CNRS, UMR 8576, UGSF, Unité de Glycobiologie Structurale et Fonctionnelle, FR 59000, Lille, France.,UGSF, Bat. C9, Université de Lille - Sciences et Technologies, 59655, Villeneuve d'Ascq, France
| | - Lucie Portier
- Univ. Lille, CNRS, UMR 8576, UGSF, Unité de Glycobiologie Structurale et Fonctionnelle, FR 59000, Lille, France.,UGSF, Bat. C9, Université de Lille - Sciences et Technologies, 59655, Villeneuve d'Ascq, France
| | - Marie-Christine Slomianny
- Univ. Lille, CNRS, UMR 8576, UGSF, Unité de Glycobiologie Structurale et Fonctionnelle, FR 59000, Lille, France.,UGSF, Bat. C9, Université de Lille - Sciences et Technologies, 59655, Villeneuve d'Ascq, France
| | - Alain Charbit
- Université Paris Descartes, Sorbonne Paris Cité, Paris, France.,Institut Necker Enfants-Malades INSERM, U1151, Team 11, Unité de Pathogénie des Infections Systémiques, Paris, France
| |
Collapse
|
48
|
Loke I, Kolarich D, Packer NH, Thaysen-Andersen M. Emerging roles of protein mannosylation in inflammation and infection. Mol Aspects Med 2016; 51:31-55. [PMID: 27086127 DOI: 10.1016/j.mam.2016.04.004] [Citation(s) in RCA: 69] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2016] [Revised: 04/05/2016] [Accepted: 04/10/2016] [Indexed: 02/07/2023]
Abstract
Proteins are frequently modified by complex carbohydrates (glycans) that play central roles in maintaining the structural and functional integrity of cells and tissues in humans and lower organisms. Mannose forms an essential building block of protein glycosylation, and its functional involvement as components of larger and diverse α-mannosidic glycoepitopes in important intra- and intercellular glycoimmunological processes is gaining recognition. With a focus on the mannose-rich asparagine (N-linked) glycosylation type, this review summarises the increasing volume of literature covering human and non-human protein mannosylation, including their structures, biosynthesis and spatiotemporal expression. The review also covers their known interactions with specialised host and microbial mannose-recognising C-type lectin receptors (mrCLRs) and antibodies (mrAbs) during inflammation and pathogen infection. Advances in molecular mapping technologies have recently revealed novel immuno-centric mannose-terminating truncated N-glycans, termed paucimannosylation, on human proteins. The cellular presentation of α-mannosidic glycoepitopes on N-glycoproteins appears tightly regulated; α-mannose determinants are relative rare glycoepitopes in physiological extracellular environments, but may be actively secreted or leaked from cells to transmit potent signals when required. Simultaneously, our understanding of the molecular basis on the recognition of mannosidic epitopes by mrCLRs including DC-SIGN, mannose receptor, mannose binding lectin and mrAb is rapidly advancing, together with the functional implications of these interactions in facilitating an effective immune response during physiological and pathophysiological conditions. Ultimately, deciphering these complex mannose-based receptor-ligand interactions at the detailed molecular level will significantly advance our understanding of immunological disorders and infectious diseases, promoting the development of future therapeutics to improve patient clinical outcomes.
Collapse
Affiliation(s)
- Ian Loke
- Department of Chemistry and Biomolecular Sciences, Macquarie University, Sydney, NSW 2109, Australia
| | - Daniel Kolarich
- Department of Biomolecular Systems, Max Planck Institute of Colloids and Interfaces, 14424 Potsdam, Germany
| | - Nicolle H Packer
- Department of Chemistry and Biomolecular Sciences, Macquarie University, Sydney, NSW 2109, Australia
| | - Morten Thaysen-Andersen
- Department of Chemistry and Biomolecular Sciences, Macquarie University, Sydney, NSW 2109, Australia.
| |
Collapse
|
49
|
Bai L, Li Q, Li L, Lin Y, Zhao S, Wang W, Wang R, Li Y, Yuan J, Wang C, Wang Z, Fan J, Liu E. Plasma High-Mannose and Complex/Hybrid N-Glycans Are Associated with Hypercholesterolemia in Humans and Rabbits. PLoS One 2016; 11:e0146982. [PMID: 26999365 PMCID: PMC4801423 DOI: 10.1371/journal.pone.0146982] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2015] [Accepted: 12/23/2015] [Indexed: 12/19/2022] Open
Abstract
N-glycans play important roles in various pathophysiological processes and can be used as clinical diagnosis markers. However, plasma N-glycans change and their pathophysiological significance in the setting of hypercholesterolemia, a major risk factor for atherosclerosis, is unknown. Here, we collected plasma from both hypercholesterolemic patients and cholesterol-fed hypercholesterolemic rabbits, and determined the changes in the whole-plasma N-glycan profile by electrospray ionization mass spectrometry. We found that both the hypercholesterolemic patients and rabbits showed a dramatic change in their plasma glycan profile. Compared with healthy subjects, the hypercholesterolemic patients exhibited higher plasma levels of a cluster of high-mannose and complex/hybrid N-glycans (mainly including undecorated or sialylated glycans), whereas only a few fucosylated or fucosylated and sialylated N-glycans were increased. Additionally, cholesterol-fed hypercholesterolemic rabbits also displayed increased plasma levels of high-mannose in addition to high complex/hybrid N-glycan levels. The whole-plasma glycan profiles revealed that the plasma N-glycan levels were correlated with the plasma cholesterol levels, implying that N-glycans may be a target for treatment of hypercholesterolemia.
Collapse
Affiliation(s)
- Liang Bai
- Research Institute of Atherosclerotic Disease, Xi’an Jiaotong University Cardiovascular Research Center, Xi’an, Shaanxi, 710061, China
- Laboratory Animal Center, Xi’an Jiaotong University Health Science Center, Xi’an, Shaanxi, 710061, China
| | - Qianwei Li
- Research Institute of Atherosclerotic Disease, Xi’an Jiaotong University Cardiovascular Research Center, Xi’an, Shaanxi, 710061, China
- Laboratory Animal Center, Xi’an Jiaotong University Health Science Center, Xi’an, Shaanxi, 710061, China
| | - Lingmei Li
- Educational Ministry Key Laboratory of Resource Biology and Biotechnology in Western China, College of Life Science, Northwest University, Xi’an, 710069, China
| | - Yan Lin
- Research Institute of Atherosclerotic Disease, Xi’an Jiaotong University Cardiovascular Research Center, Xi’an, Shaanxi, 710061, China
- Laboratory Animal Center, Xi’an Jiaotong University Health Science Center, Xi’an, Shaanxi, 710061, China
| | - Sihai Zhao
- Research Institute of Atherosclerotic Disease, Xi’an Jiaotong University Cardiovascular Research Center, Xi’an, Shaanxi, 710061, China
- Laboratory Animal Center, Xi’an Jiaotong University Health Science Center, Xi’an, Shaanxi, 710061, China
| | - Weirong Wang
- Research Institute of Atherosclerotic Disease, Xi’an Jiaotong University Cardiovascular Research Center, Xi’an, Shaanxi, 710061, China
- Laboratory Animal Center, Xi’an Jiaotong University Health Science Center, Xi’an, Shaanxi, 710061, China
| | - Rong Wang
- Research Institute of Atherosclerotic Disease, Xi’an Jiaotong University Cardiovascular Research Center, Xi’an, Shaanxi, 710061, China
- Laboratory Animal Center, Xi’an Jiaotong University Health Science Center, Xi’an, Shaanxi, 710061, China
| | - Yongqin Li
- Department of Cardiology, the Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi, 710004, China
| | - Jiangbei Yuan
- Educational Ministry Key Laboratory of Resource Biology and Biotechnology in Western China, College of Life Science, Northwest University, Xi’an, 710069, China
| | - Chengjian Wang
- Educational Ministry Key Laboratory of Resource Biology and Biotechnology in Western China, College of Life Science, Northwest University, Xi’an, 710069, China
| | - Zhongfu Wang
- Educational Ministry Key Laboratory of Resource Biology and Biotechnology in Western China, College of Life Science, Northwest University, Xi’an, 710069, China
| | - Jianglin Fan
- Department of Molecular Pathology, Interdisciplinary Graduate School of Medicine and Engineering, University of Yamanashi, Yamanashi, 409–3898, Japan
| | - Enqi Liu
- Research Institute of Atherosclerotic Disease, Xi’an Jiaotong University Cardiovascular Research Center, Xi’an, Shaanxi, 710061, China
- Laboratory Animal Center, Xi’an Jiaotong University Health Science Center, Xi’an, Shaanxi, 710061, China
| |
Collapse
|
50
|
Niang B, Jin L, Chen X, Guo X, Zhang H, Wu Q, Padhiar AA, Xiao M, Fang D, Zhang J. GalNAc-T4 putatively modulates the estrogen regulatory network through FOXA1 glycosylation in human breast cancer cells. Mol Cell Biochem 2016; 411:393-402. [PMID: 26541755 DOI: 10.1007/s11010-015-2601-1] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2015] [Accepted: 10/29/2015] [Indexed: 12/31/2022]
Abstract
GALNT4 belongs to a family of N-acetylgalactosaminyltransferases, which catalyze the transfer of GalNAc to Serine or Threonine residues in the initial step of mucin-type O-linked protein glycosylation. This glycosylation type is the most complex post-translational modification of proteins, playing important roles during cellular differentiation and in pathological disorders. Most of the breast cancer subtypes are estrogen receptor positive, and hence, the estrogen pathway represents a key regulatory network. We investigated the expression of GalNAc-T4 in a panel of mammary epithelial cell lines and found its expression is associated with the estrogen status of the cells. FOXA1, a key transcription factor, functions to promote estrogen responsive gene expression by acting as a cofactor to estrogen receptor alpha (ERα), but all the aspects of this regulatory mechanism are not fully explored. This study found that knockdown of GALNT4 expression in human breast cancer cells attenuated the protein expression of ERα, FOXA1, and Cyclin D1. Further, our immunoprecipitation assays depicted the possibility of FOXA1 to undergo O-GalNAc modifications with a decrease of GalNAc residues in the GALNT4 knockdown cells and also impairment in the FOXA1-ERα association. Rescuing GALNT4 expression could restore the interaction as well as the glycosylation of FOXA1. Together, these findings suggest a key role for GalNAc-T4 in the estrogen pathway through FOXA1 glycosylation.
Collapse
Affiliation(s)
- Bachir Niang
- Department of Biochemistry, Institute of Glycobiology, Dalian Medical University, 9 South Lvshun Road Western Section, Dalian, 116044, China
| | - Liyuan Jin
- Department of Biochemistry, Institute of Glycobiology, Dalian Medical University, 9 South Lvshun Road Western Section, Dalian, 116044, China
| | - Xixi Chen
- Department of Biochemistry, Institute of Glycobiology, Dalian Medical University, 9 South Lvshun Road Western Section, Dalian, 116044, China
| | - Xiaohan Guo
- Department of Biochemistry, Institute of Glycobiology, Dalian Medical University, 9 South Lvshun Road Western Section, Dalian, 116044, China
| | - Hongshuo Zhang
- Department of Biochemistry, Institute of Glycobiology, Dalian Medical University, 9 South Lvshun Road Western Section, Dalian, 116044, China
| | - Qiong Wu
- School of Life Science and Medicine, Dalian University of Technology, 2 Linggong Road, Dalian, 116024, China
| | - Arshad Ahmed Padhiar
- Department of Biochemistry, Institute of Glycobiology, Dalian Medical University, 9 South Lvshun Road Western Section, Dalian, 116044, China
| | - Min Xiao
- National Glycoengineering Research Center and State Key Laboratory of Microbial Technology, Shandong University, Jinan, 250100, China
| | - Deyu Fang
- Department of Pathology, Northwestern University Feinberg School of Medicine, 303 E. Chicago Ave, Chicago, IL, 60611, USA
| | - Jianing Zhang
- Department of Biochemistry, Institute of Glycobiology, Dalian Medical University, 9 South Lvshun Road Western Section, Dalian, 116044, China.
- School of Life Science and Medicine, Dalian University of Technology, 2 Linggong Road, Dalian, 116024, China.
| |
Collapse
|