1
|
Qiu Y, Gao S, Ding X, Lu J, Ji X, Hao W, Cheng S, Du H, Gu Y, Yu C, Cheng C, Gao X. Conditional Tnfaip6-Knockout in Inner Ear Hair Cells Does not Alter Auditory Function. Neurosci Bull 2024:10.1007/s12264-024-01326-8. [PMID: 39688649 DOI: 10.1007/s12264-024-01326-8] [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: 02/23/2024] [Accepted: 06/05/2024] [Indexed: 12/18/2024] Open
Abstract
Noise-induced hearing loss is a worldwide public health issue that is characterized by temporary or permanent changes in hearing sensitivity. This condition is closely linked to inflammatory responses, and interventions targeting the inflammatory gene tumor necrosis factor-alpha (TNFα) are known to mitigate cochlear noise damage. TNFα-induced proteins (TNFAIPs) are a family of translucent acidic proteins, and TNFAIP6 has a notable association with inflammatory responses. To date, there have been few reports on TNFAIP6 levels in the inner ear. To elucidate the precise mechanism, we generated transgenic mouse models with conditional knockout of Tnfaip6 (Tnfaip6 cKO). Evaluation of hair cell morphology and function revealed no significant differences in hair cell numbers or ribbon synapses between Tnfaip6 cKO and wild-type mice. Moreover, there were no notable variations in hair cell numbers or hearing function in noisy environments. Our results indicate that Tnfaip6 does not have a substantial impact on the auditory system.
Collapse
Affiliation(s)
- Yue Qiu
- Department of Otolaryngology-Head and Neck Surgery, Jiangsu Provincial Key Medical Discipline (Laboratory), Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, 210008, China
| | - Song Gao
- Department of Otolaryngology-Head and Neck Surgery, Jiangsu Provincial Key Medical Discipline (Laboratory), Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, 210008, China
| | - Xiaoqiong Ding
- Department of Otolaryngology-Head and Neck Surgery, Zhongda Hospital, Southeast University, Nanjing, 210008, China
| | - Jie Lu
- Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Yangzhou, 225001, China
| | - Xinya Ji
- Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Yangzhou, 225001, China
| | - Wenli Hao
- Department of Otolaryngology-Head and Neck Surgery, Jiangsu Provincial Key Medical Discipline (Laboratory), Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, 210008, China
| | - Siqi Cheng
- Department of Otolaryngology-Head and Neck Surgery, Jiangsu Provincial Key Medical Discipline (Laboratory), Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, 210008, China
| | - Haolinag Du
- Department of Otolaryngology-Head and Neck Surgery, Jiangsu Provincial Key Medical Discipline (Laboratory), Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, 210008, China
| | - Yajun Gu
- Department of Otolaryngology-Head and Neck Surgery, Jiangsu Provincial Key Medical Discipline (Laboratory), Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, 210008, China
| | - Chenjie Yu
- Department of Otolaryngology-Head and Neck Surgery, Jiangsu Provincial Key Medical Discipline (Laboratory), Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, 210008, China.
- Research Institute of Otolaryngology, Nanjing, 210008, China.
| | - Cheng Cheng
- Department of Otolaryngology-Head and Neck Surgery, Jiangsu Provincial Key Medical Discipline (Laboratory), Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, 210008, China.
- Research Institute of Otolaryngology, Nanjing, 210008, China.
| | - Xia Gao
- Department of Otolaryngology-Head and Neck Surgery, Jiangsu Provincial Key Medical Discipline (Laboratory), Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, 210008, China.
- Research Institute of Otolaryngology, Nanjing, 210008, China.
| |
Collapse
|
2
|
Tang F, Reeves SR, Brune JE, Chang MY, Chan CK, Waldron P, Drummond SP, Milner CM, Alonge KM, Garantziotis S, Day AJ, Altemeier WA, Frevert CW. Inter-alpha-trypsin inhibitor (IαI) and hyaluronan modifications enhance the innate immune response to influenza virus in the lung. Matrix Biol 2024; 126:25-42. [PMID: 38232913 DOI: 10.1016/j.matbio.2024.01.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2023] [Revised: 12/22/2023] [Accepted: 01/14/2024] [Indexed: 01/19/2024]
Abstract
The inter-alpha-trypsin inhibitor (IαI) complex is composed of the bikunin core protein with a single chondroitin sulfate (CS) attached and one or two heavy chains (HCs) covalently linked to the CS chain. The HCs from IαI can be transferred to hyaluronan (HA) through a TNFα-stimulated gene-6 (TSG-6) dependent process to form an HC•HA matrix. Previous studies reported increased IαI, HA, and HC•HA complexes in mouse bronchoalveolar lavage fluid (BALF) post-influenza infection. However, the expression and incorporation of HCs into the HA matrix of the lungs during the clinical course of influenza A virus (IAV) infection and the biological significance of the HC•HA matrix are poorly understood. The present study aimed to better understand the composition of HC•HA matrices in mice infected with IAV and how these matrices regulate the host pulmonary immune response. In IAV infected mice bikunin, HC1-3, TSG-6, and HAS1-3 all show increased gene expression at various times during a 12-day clinical course. The increased accumulation of IαI and HA was confirmed in the lungs of infected mice using immunohistochemistry and quantitative digital pathology. Western blots confirmed increases in the IαI components in BALF and lung tissue at 6 days post-infection (dpi). Interestingly, HCs and bikunin recovered from BALF and plasma from mice 6 dpi with IAV, displayed differences in the HC composition by Western blot analysis and differences in bikunin's CS chain sulfation patterns by mass spectrometry analysis. This strongly suggests that the IαI components were synthesized in the lungs rather than translocated from the vascular compartment. HA was significantly increased in BALF at 6 dpi, and the HA recovered in BALF and lung tissues were modified with HCs indicating the presence of an HC•HA matrix. In vitro experiments using polyinosinic-polycytidylic acid (poly(I:C)) treated mouse lung fibroblasts (MLF) showed that modification of HA with HCs increased cell-associated HA, and that this increase was due to the retention of HA in the MLF glycocalyx. In vitro studies of leukocyte adhesion showed differential binding of lymphoid (Hut78), monocyte (U937), and neutrophil (dHL60) cell lines to HA and HC•HA matrices. Hut78 cells adhered to immobilized HA in a size and concentration-dependent manner. In contrast, the binding of dHL60 and U937 cells depended on generating a HC•HA matrix by MLF. Our in vivo findings, using multiple bronchoalveolar lavages, correlated with our in vitro findings in that lymphoid cells bound more tightly to the HA-glycocalyx in the lungs of influenza-infected mice than neutrophils and mononuclear phagocytes (MNPs). The neutrophils and MNPs were associated with a HC•HA matrix and were more readily lavaged from the lungs. In conclusion, this work shows increased IαI and HA accumulation and the formation of a HC•HA matrix in mouse lungs post-IAV infection. The formation of HA and HC•HA matrices could potentially create specific microenvironments in the lungs for immune cell recruitment and activation during IAV infection.
Collapse
Affiliation(s)
- Fengying Tang
- Center for Lung Biology, the University of Washington at South Lake Union, Seattle, WA, USA; Department of Comparative Medicine, University of Washington, Seattle, WA, USA.
| | - Stephen R Reeves
- Center for Respiratory Biology and Therapeutics, Seattle Children's Research Institute, Seattle, WA, USA; Division of Pulmonary and Sleep Medicine, Department of Pediatrics, University of Washington, Seattle, WA, USA
| | - Jourdan E Brune
- Center for Lung Biology, the University of Washington at South Lake Union, Seattle, WA, USA; Department of Comparative Medicine, University of Washington, Seattle, WA, USA
| | - Mary Y Chang
- Center for Lung Biology, the University of Washington at South Lake Union, Seattle, WA, USA; Department of Comparative Medicine, University of Washington, Seattle, WA, USA
| | - Christina K Chan
- Center for Lung Biology, the University of Washington at South Lake Union, Seattle, WA, USA; Department of Comparative Medicine, University of Washington, Seattle, WA, USA
| | - Peter Waldron
- Center for Lung Biology, the University of Washington at South Lake Union, Seattle, WA, USA; Department of Comparative Medicine, University of Washington, Seattle, WA, USA
| | - Sheona P Drummond
- Welcome Centre for Cell-Matrix Research, University of Manchester, Manchester, UK; Faculty of Biology Medicine & Health, University of Manchester, Manchester Academic Health Science Centre, Manchester, UK
| | - Caroline M Milner
- Faculty of Biology Medicine & Health, University of Manchester, Manchester Academic Health Science Centre, Manchester, UK; Lydia Becker Institute of Immunology and Inflammation, University of Manchester, Manchester, UK
| | - Kimberly M Alonge
- Department of Medicinal Chemistry, University of Washington, Seattle, WA, USA
| | - Stavros Garantziotis
- Division of Intramural Research, National Institute of Environmental Health Sciences, Research Triangle Park, NC, USA
| | - Anthony J Day
- Welcome Centre for Cell-Matrix Research, University of Manchester, Manchester, UK; Faculty of Biology Medicine & Health, University of Manchester, Manchester Academic Health Science Centre, Manchester, UK; Lydia Becker Institute of Immunology and Inflammation, University of Manchester, Manchester, UK
| | - William A Altemeier
- Center for Lung Biology, the University of Washington at South Lake Union, Seattle, WA, USA; Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, University of Washington, Seattle, WA, USA
| | - Charles W Frevert
- Center for Lung Biology, the University of Washington at South Lake Union, Seattle, WA, USA; Department of Comparative Medicine, University of Washington, Seattle, WA, USA; Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, University of Washington, Seattle, WA, USA
| |
Collapse
|
3
|
Noborn F, Nilsson J, Sihlbom C, Nikpour M, Kjellén L, Larson G. Mapping the Human Chondroitin Sulfate Glycoproteome Reveals an Unexpected Correlation Between Glycan Sulfation and Attachment Site Characteristics. Mol Cell Proteomics 2023; 22:100617. [PMID: 37453717 PMCID: PMC10424144 DOI: 10.1016/j.mcpro.2023.100617] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Revised: 07/07/2023] [Accepted: 07/12/2023] [Indexed: 07/18/2023] Open
Abstract
Chondroitin sulfate proteoglycans (CSPGs) control key events in human health and disease and are composed of chondroitin sulfate (CS) polysaccharide(s) attached to different core proteins. Detailed information on the biological effects of site-specific CS structures is scarce as the polysaccharides are typically released from their core proteins prior to analysis. Here we present a novel glycoproteomic approach for site-specific sequencing of CS modifications from human urine. Software-assisted and manual analysis revealed that certain core proteins carried CS with abundant sulfate modifications, while others carried CS with lower levels of sulfation. Inspection of the amino acid sequences surrounding the attachment sites indicated that the acidity of the attachment site motifs increased the levels of CS sulfation, and statistical analysis confirmed this relationship. However, not only the acidity but also the sequence and characteristics of specific amino acids in the proximity of the serine glycosylation site correlated with the degree of sulfation. These results demonstrate attachment site-specific characteristics of CS polysaccharides of CSPGs in human urine and indicate that this novel method may assist in elucidating the biosynthesis and functional roles of CSPGs in cellular physiology.
Collapse
Affiliation(s)
- Fredrik Noborn
- Department of Laboratory Medicine, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Jonas Nilsson
- Proteomics Core Facility, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Carina Sihlbom
- Proteomics Core Facility, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Mahnaz Nikpour
- Department of Laboratory Medicine, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Lena Kjellén
- Department of Medical Biochemistry and Microbiology, Uppsala University, Uppsala, Sweden
| | - Göran Larson
- Department of Laboratory Medicine, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden; Laboratory of Clinical Chemistry, Sahlgrenska University Hospital, Gothenburg, Sweden.
| |
Collapse
|
4
|
Lepedda AJ, Nieddu G, Cannas C, Formato M. Molecular and pathobiological insights of bikunin/UTI in cancer. Mol Biol Rep 2023; 50:1701-1711. [PMID: 36414878 PMCID: PMC9889512 DOI: 10.1007/s11033-022-08117-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Accepted: 11/12/2022] [Indexed: 11/24/2022]
Abstract
Bikunin is a small chondroitin sulfate proteoglycan (PG) with Ser-protease inhibitory activity that plays pleiotropic roles in health and disease. It is involved in several physiological processes including stabilization of the extracellular matrix (ECM) of connective tissues and key reproductive events. Bikunin is also implicated in both acute and chronic inflammatory conditions and represents a non-invasive circulating and/or urinary (as Urinary Trypsin Inhibitor or UTI) biomarker. It exerts inhibitory effects on urokinase-type plasminogen activator (uPA) and its receptor (uPAR) mediating tumor invasiveness by a down-regulation of uPA mRNA expression, thus representing an anti-metastatic agent. However, only limited data on its potential as a diagnostic and/or prognostic marker of cancer have been reported so far. Recent technological advances in mass spectrometry-based proteomics have provided researchers with a huge amount of information allowing for large-scale surveys of the cancer proteome. To address such issues, we analyzed bikunin expression data across several types of tumors, by using UALCAN proteogenomic analysis portal. In this article we critically review the roles of bikunin in human pathobiology, with a special focus on its inhibitory effects and mechanisms in cancer aggressiveness as well as its significance as cancer circulating biomarker.
Collapse
Affiliation(s)
| | - Gabriele Nieddu
- Department of Biomedical Sciences, University of Sassari, Sassari, Italy
| | - Claudia Cannas
- Department of Biomedical Sciences, University of Sassari, Sassari, Italy
| | - Marilena Formato
- Department of Biomedical Sciences, University of Sassari, Sassari, Italy
| |
Collapse
|
5
|
Ning Y, Zhang P, Zhang F, Chen S, Liu Y, Chen F, Wu Y, Li S, Wang C, Gong Y, Hu M, Huang R, Zhao H, Guo X, Wang X, Yang L. Abnormal expression of TSG-6 disturbs extracellular matrix homeostasis in chondrocytes from endemic osteoarthritis. Front Genet 2022; 13:1064565. [DOI: 10.3389/fgene.2022.1064565] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2022] [Accepted: 11/02/2022] [Indexed: 11/19/2022] Open
Abstract
Background and aims: Kashin-Beck disease (KBD) is a unique endemic osteochondropathy with unclear pathogenesis in China. T-2 toxin exposure has been identified as a significant risk factor of KBD. However, the mechanism of articular cartilage damage induced by T-2 toxin is a conundrum. We explored the role of the extracellular matrix-related gene TSG-6 in the articular chondrocyte damage process under the exposure of HT-2 toxin.Methods: TSG-6 was identified as a candidate gene by mining our previous gene expression profiling of KBD and verified by qRT-PCR and immunohistochemistry. Then, TSG-6 was silenced by RNA interference technology and overexpressed induction by TNF-α. Gradient concentrations of HT-2 toxin were added to intervene with C28/I2 chondrocytes. MTT was used to observe the proliferation and cell viability of chondrocytes, and qRT-PCR was utilized to detect the expression changes of MMP1, MMP3, MMP13, COL2A1, and proteoglycan before and after treatments for verification.Results: TSG-6 was upregulated in KBD chondrocytes at the mRNA level and upregulated in the superficial, middle, and deep zones of KBD cartilage. After TSG-6 silencing, the expression of MMP1, MMP3, MMP13, and proteoglycan was significantly decreased while COL2A1 expression was significantly increased, which was reversed after the overexpression of TSG-6 induced by TNF-α (p < 0.05). The survival rate of chondrocytes was correspondingly reduced with an increase in the HT-2 toxin concentration. Compared with the blank control group, the expression of MMPs was increased in the intervention group of HT-2 toxin, while the expression of proteoglycan and COL2A1 decreased (p < 0.05).Conclusion: The upregulation of the TSG-6 gene may play a role in promoting the damage and degradation of the extracellular matrix in KBD chondrocytes under the exposure of HT-2 toxin.
Collapse
|
6
|
Gatto F, Dabestani S, Bratulic S, Limeta A, Maccari F, Galeotti F, Volpi N, Stierner U, Nielsen J, Lundstam S. Plasma and Urine Free Glycosaminoglycans as Monitoring Biomarkers in Nonmetastatic Renal Cell Carcinoma-A Prospective Cohort Study. EUR UROL SUPPL 2022; 42:30-39. [PMID: 35911082 PMCID: PMC9334826 DOI: 10.1016/j.euros.2022.06.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/09/2022] [Indexed: 11/17/2022] Open
Abstract
Background No liquid biomarkers are approved in renal cell carcinoma (RCC), making early detection of recurrence in surgically treated nonmetastatic (M0) patients dependent on radiological imaging. Urine- and plasma free glycosaminoglycan profiles-or free GAGomes-are promising biomarkers reflective of RCC metabolism. Objective To explore whether free GAGomes could detect M0 RCC recurrence noninvasively. Design setting and participants Between June 2016 and February 2021, we enrolled a prospective consecutive series of patients elected for (1) partial or radical nephrectomy for clinical M0 RCC (cohort 1) or (2) first-line therapy following RCC metachronous metastatic recurrence (cohort 2) at Sahlgrenska University Hospital, Gothenburg, Sweden. The study population included M0 RCC patients with recurrent disease (RD) versus no evidence of disease (NED) in at least one follow-up visit. Plasma and urine free GAGomes-consisting of 40 chondroitin sulfate (CS), heparan sulfate, and hyaluronic acid (HA) features-were measured in a blinded central laboratory preoperatively and at each postoperative follow-up visit until recurrence or end of follow-up in cohort 1, or before treatment start in cohort 2. Outcome measurements and statistical analysis We used Bayesian logistic regression to correlate GAGome features with RD versus NED and with various histopathological variables. We developed three recurrence scores (plasma, urine, and combined) proportional to the predicted probability of RD. We internally validated the area under the curve (AUC) using bootstrap resampling. We performed a decision curve analysis to select a cutoff and report the corresponding net benefit, sensitivity, and specificity of each score. We used univariable analyses to correlate each preoperative score with recurrence-free survival (RFS). Results and limitations Of 127 enrolled patients in total, 62 M0 RCC patients were in the study population (median age: 63 year, 35% female, and 82% clear cell). The median follow-up time was 3 months, totaling 72 postoperative visits -17 RD and 55 NED cases. RD was compatible with alterations in 14 (52%) of the detectable GAGome features, mostly free CS. Eleven (79%) of these correlated with at least one histopathological variable. We developed a plasma, a urine, and a combined free CS RCC recurrence score to diagnose RD versus NED with AUCs 0.91, 0.93, and 0.94, respectively. At a cutoff equivalent to ≥30% predicted probability of RD, the sensitivity and specificity were, respectively, 69% and 84% in plasma, 81% and 80% in urine, and 80% and 82% when combined, and the net benefit was equivalent to finding an extra ten, 13, and 12 cases of RD per hundred patients without any unnecessary imaging for plasma, urine, and combined, respectively. The combined score was prognostic of RFS in univariable analysis (hazard ratio = 1.90, p = 0.02). Limitations include a lack of external validation. Conclusions Free CS scores detected postsurgical recurrence noninvasively in M0 RCC with substantial net benefit. External validity is required before wider clinical implementation. Patient summary In this study, we examined a new noninvasive blood and urine test to detect whether renal cell carcinoma recurred after surgery.
Collapse
Affiliation(s)
- Francesco Gatto
- Department of Biology and Biological Engineering, Chalmers University of Technology, Gothenburg, Sweden
| | - Saeed Dabestani
- Department of Translational Medicine, Division of Urological Cancers, Lund University, Kristianstad Central Hospital, Region Skane, Lund, Sweden
- Department of Urology, Kristianstad Central Hospital, Region Skane, Kristianstad, Sweden
| | - Sinisa Bratulic
- Department of Biology and Biological Engineering, Chalmers University of Technology, Gothenburg, Sweden
| | - Angelo Limeta
- Department of Biology and Biological Engineering, Chalmers University of Technology, Gothenburg, Sweden
| | - Francesca Maccari
- Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Fabio Galeotti
- Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Nicola Volpi
- Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Ulrika Stierner
- Department of Oncology, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Jens Nielsen
- Department of Biology and Biological Engineering, Chalmers University of Technology, Gothenburg, Sweden
- BioInnovation Institute, Copenhagen N, Denmark
| | - Sven Lundstam
- Department of Oncology, Sahlgrenska University Hospital, Gothenburg, Sweden
- Department of Urology, Sahlgrenska University Hospital, Gothenburg, Sweden
| |
Collapse
|
7
|
Long noncoding RNA LINC00261 upregulates ITIH5 to impair tumorigenic ability of pancreatic cancer stem cells. Cell Death Discov 2021; 7:220. [PMID: 34446696 PMCID: PMC8390744 DOI: 10.1038/s41420-021-00575-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Revised: 03/25/2021] [Accepted: 04/23/2021] [Indexed: 12/11/2022] Open
Abstract
Long noncoding RNAs (lncRNAs) are implicated tumor development in a range of different cancers, including pancreatic cancer (PC). Cancer stem cells (CSCs), a drug-resistant cancer cell subset, drive tumor progression in PC. In this work, we aimed to investigate the mechanism by which lncRNA LINC00261 affects the biological functions of CSCs during the progression of PC. Microarray analysis of differentially expressed genes and lncRNAs suggested that LINC00261 is downregulated in PC. Both LINC00261 and ITIH5 were confirmed to be downregulated in PC cells and PC stem cells. Gain-of-function and loss-of-function investigations were performed to analyze their effects on cell proliferation, drug resistance, cell cycle distribution, self-renewal, invasion, and ultimately overall tumorigenicity. These experiments revealed that the expression of stem cell markers was reduced, and cell proliferation, self-renewal ability, cell invasion, drug resistance, and tumorigenicity were all suppressed by upregulation of LINC00261 or ITIH5. The results of dual-luciferase reporter gene, ChIP, and RIP assays indicated that LINC00261 binds directly to GATA6, increasing its activity at the ITIH5 promoter. The presence of LINC00261 and GATA6 inhibited the self-renewal and tumorigenesis of PC stem cells, while silence of ITIH5 rescued those functions. Collectively, this study identifies the tumor suppressive activity of LINC00261 in PC, showing that this lncRNA limits the functions of PC stem through an ITIH5/GATA6 regulatory pathway.
Collapse
|
8
|
Noborn F, Nikpour M, Persson A, Nilsson J, Larson G. Expanding the Chondroitin Sulfate Glycoproteome - But How Far? Front Cell Dev Biol 2021; 9:695970. [PMID: 34490248 PMCID: PMC8418075 DOI: 10.3389/fcell.2021.695970] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Accepted: 07/27/2021] [Indexed: 12/15/2022] Open
Abstract
Chondroitin sulfate proteoglycans (CSPGs) are found at cell surfaces and in connective tissues, where they interact with a multitude of proteins involved in various pathophysiological processes. From a methodological perspective, the identification of CSPGs is challenging, as the identification requires the combined sequencing of specific core proteins, together with the characterization of the CS polysaccharide modification(s). According to the current notion of CSPGs, they are often considered in relation to a functional role in which a given proteoglycan regulates a specific function in cellular physiology. Recent advances in glycoproteomic methods have, however, enabled the identification of numerous novel chondroitin sulfate core proteins, and their glycosaminoglycan attachment sites, in humans and in various animal models. In addition, these methods have revealed unexpected structural complexity even in the linkage regions. These findings indicate that the number and structural complexity of CSPGs are much greater than previously perceived. In light of these findings, the prospect of finding additional CSPGs, using improved methods for structural and functional characterizations, and studying novel sample matrices in humans and in animal models is discussed. Further, as many of the novel CSPGs are found in low abundance and with not yet assigned functions, these findings may challenge the traditional notion of defining proteoglycans. Therefore, the concept of proteoglycans is considered, discussing whether "a proteoglycan" should be defined mainly on the basis of an assigned function or on the structural evidence of its existence.
Collapse
Affiliation(s)
- Fredrik Noborn
- Department of Laboratory Medicine, Institute of Biomedicine, Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - Mahnaz Nikpour
- Department of Laboratory Medicine, Institute of Biomedicine, Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - Andrea Persson
- Department of Laboratory Medicine, Institute of Biomedicine, Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - Jonas Nilsson
- Department of Laboratory Medicine, Institute of Biomedicine, Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
- Proteomics Core Facility, Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - Göran Larson
- Department of Laboratory Medicine, Institute of Biomedicine, Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| |
Collapse
|
9
|
Hayes AJ, Melrose J. Neural Tissue Homeostasis and Repair Is Regulated via CS and DS Proteoglycan Motifs. Front Cell Dev Biol 2021; 9:696640. [PMID: 34409033 PMCID: PMC8365427 DOI: 10.3389/fcell.2021.696640] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Accepted: 07/13/2021] [Indexed: 01/04/2023] Open
Abstract
Chondroitin sulfate (CS) is the most abundant and widely distributed glycosaminoglycan (GAG) in the human body. As a component of proteoglycans (PGs) it has numerous roles in matrix stabilization and cellular regulation. This chapter highlights the roles of CS and CS-PGs in the central and peripheral nervous systems (CNS/PNS). CS has specific cell regulatory roles that control tissue function and homeostasis. The CNS/PNS contains a diverse range of CS-PGs which direct the development of embryonic neural axonal networks, and the responses of neural cell populations in mature tissues to traumatic injury. Following brain trauma and spinal cord injury, a stabilizing CS-PG-rich scar tissue is laid down at the defect site to protect neural tissues, which are amongst the softest tissues of the human body. Unfortunately, the CS concentrated in gliotic scars also inhibits neural outgrowth and functional recovery. CS has well known inhibitory properties over neural behavior, and animal models of CNS/PNS injury have demonstrated that selective degradation of CS using chondroitinase improves neuronal functional recovery. CS-PGs are present diffusely in the CNS but also form denser regions of extracellular matrix termed perineuronal nets which surround neurons. Hyaluronan is immobilized in hyalectan CS-PG aggregates in these perineural structures, which provide neural protection, synapse, and neural plasticity, and have roles in memory and cognitive learning. Despite the generally inhibitory cues delivered by CS-A and CS-C, some CS-PGs containing highly charged CS disaccharides (CS-D, CS-E) or dermatan sulfate (DS) disaccharides that promote neural outgrowth and functional recovery. CS/DS thus has varied cell regulatory properties and structural ECM supportive roles in the CNS/PNS depending on the glycoform present and its location in tissue niches and specific cellular contexts. Studies on the fruit fly, Drosophila melanogaster and the nematode Caenorhabditis elegans have provided insightful information on neural interconnectivity and the role of the ECM and its PGs in neural development and in tissue morphogenesis in a whole organism environment.
Collapse
Affiliation(s)
- Anthony J. Hayes
- Bioimaging Research Hub, Cardiff School of Biosciences, Cardiff University, Wales, United Kingdom
| | - James Melrose
- Graduate School of Biomedical Engineering, University of New South Wales, Sydney, NSW, Australia
- Raymond Purves Bone and Joint Research Laboratories, Kolling Institute of Medical Research, Royal North Shore Hospital and The Faculty of Medicine and Health, The University of Sydney, St. Leonard’s, NSW, Australia
| |
Collapse
|
10
|
Chandrasekar K, Farrugia BL, Johnson L, Marks D, Irving D, Elgundi Z, Lau K, Kim HN, Rnjak‐Kovacina J, Bilek MM, Whitelock JM, Lord MS. Effect of Recombinant Human Perlecan Domain V Tethering Method on Protein Orientation and Blood Contacting Activity on Polyvinyl Chloride. Adv Healthc Mater 2021; 10:e2100388. [PMID: 33890424 DOI: 10.1002/adhm.202100388] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Revised: 04/06/2021] [Indexed: 12/23/2022]
Abstract
Surface modification of biomaterials is a promising approach to control biofunctionality while retaining the bulk biomaterial properties. Perlecan is the major proteoglycan in the vascular basement membrane that supports low levels of platelet adhesion but not activation. Thus, perlecan is a promising bioactive for blood-contacting applications. This study furthers the mechanistic understanding of platelet interactions with perlecan by establishing that platelets utilize domains III and V of the core protein for adhesion. Polyvinyl chloride (PVC) is functionalized with recombinant human perlecan domain V (rDV) to explore the effect of the tethering method on proteoglycan orientation and bioactivity. Tethering of rDV to PVC is achieved via either physisorption or covalent attachment via plasma immersion ion implantation (PIII) treatment. Both methods of rDV tethering reduce platelet adhesion and activation compared to the pristine PVC, however, the mechanisms are unique for each tethering method. Physisorption of rDV on PVC orientates the molecule to hinder access to the integrin-binding region, which inhibits platelet adhesion. In contrast, PIII treatment orientates rDV to allow access to the integrin-binding region, which is rendered antiadhesive to platelets via the glycosaminoglycan (GAG) chain. These effects demonstrate the potential of rDV biofunctionalization to modulate platelet interactions for blood contacting applications.
Collapse
Affiliation(s)
| | - Brooke L. Farrugia
- Department of Biomedical Engineering Melbourne School of Engineering The University of Melbourne Melbourne VIC 3010 Australia
| | - Lacey Johnson
- Australian Red Cross Lifeblood Alexandria NSW 2015 Australia
| | - Denese Marks
- Australian Red Cross Lifeblood Alexandria NSW 2015 Australia
| | - David Irving
- Australian Red Cross Lifeblood Alexandria NSW 2015 Australia
| | - Zehra Elgundi
- Graduate School of Biomedical Engineering UNSW Sydney Sydney NSW 2052 Australia
| | - Kieran Lau
- Graduate School of Biomedical Engineering UNSW Sydney Sydney NSW 2052 Australia
| | - Ha Na Kim
- Graduate School of Biomedical Engineering UNSW Sydney Sydney NSW 2052 Australia
| | | | - Marcela M. Bilek
- The Charles Perkins Centre University of Sydney Sydney NSW 2006 Australia
- The University of Sydney Nano Institute University of Sydney Sydney NSW 2006 Australia
- School of Physics University of Sydney Sydney NSW 2006 Australia
- School of Biomedical Engineering University of Sydney Sydney NSW 2006 Australia
| | - John M. Whitelock
- Graduate School of Biomedical Engineering UNSW Sydney Sydney NSW 2052 Australia
| | - Megan S. Lord
- Graduate School of Biomedical Engineering UNSW Sydney Sydney NSW 2052 Australia
| |
Collapse
|
11
|
Lettow M, Greis K, Grabarics M, Horlebein J, Miller RL, Meijer G, von Helden G, Pagel K. Chondroitin Sulfate Disaccharides in the Gas Phase: Differentiation and Conformational Constraints. J Phys Chem A 2021; 125:4373-4379. [PMID: 33979516 PMCID: PMC8279649 DOI: 10.1021/acs.jpca.1c02463] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
![]()
Glycosaminoglycans
(GAGs) are a family of complex carbohydrates
vital to all mammalian organisms and involved in numerous biological
processes. Chondroitin and dermatan sulfate, an important class of
GAGs, are linear macromolecules consisting of disaccharide building
blocks of N-acetylgalactosamine and two different
uronic acids. The varying degree and the site of sulfation render
their characterization challenging. Here, we combine mass spectrometry
with cryogenic infrared spectroscopy in the wavenumber range from
1000 to 1800 cm–1. Fingerprint spectra were recorded
for a comprehensive set of disaccharides bearing all known motifs
of sulfation. In addition, state-of-the-art quantum chemical calculations
were performed to aid the understanding of the differences in the
experimental fingerprint spectra. The results show that the degree
and position of charged sulfate groups define the size of the conformational
landscape in the gas phase. The detailed understanding of cryogenic
infrared spectroscopy for acidic and often highly sulfated glycans
may pave the way to utilize the technique in fragment-based sequencing
approaches.
Collapse
Affiliation(s)
- Maike Lettow
- Fritz-Haber-Institut der Max-Planck-Gesellschaft, Faradayweg 4-6, 14195 Berlin, Germany.,Institut für Chemie und Biochemie, Freie Universität Berlin, Arnimallee 22, 14195 Berlin, Germany
| | - Kim Greis
- Fritz-Haber-Institut der Max-Planck-Gesellschaft, Faradayweg 4-6, 14195 Berlin, Germany.,Institut für Chemie und Biochemie, Freie Universität Berlin, Arnimallee 22, 14195 Berlin, Germany
| | - Márkó Grabarics
- Fritz-Haber-Institut der Max-Planck-Gesellschaft, Faradayweg 4-6, 14195 Berlin, Germany.,Institut für Chemie und Biochemie, Freie Universität Berlin, Arnimallee 22, 14195 Berlin, Germany
| | - Jan Horlebein
- Fritz-Haber-Institut der Max-Planck-Gesellschaft, Faradayweg 4-6, 14195 Berlin, Germany.,Institut für Chemie und Biochemie, Freie Universität Berlin, Arnimallee 22, 14195 Berlin, Germany
| | - Rebecca L Miller
- Copenhagen Center for Glycomics, Department of Cellular and Molecular Medicine, Faculty of Health Sciences, University of Copenhagen, Blegdamsvej 3, DK-2200 Copenhagen N, Denmark
| | - Gerard Meijer
- Fritz-Haber-Institut der Max-Planck-Gesellschaft, Faradayweg 4-6, 14195 Berlin, Germany
| | - Gert von Helden
- Fritz-Haber-Institut der Max-Planck-Gesellschaft, Faradayweg 4-6, 14195 Berlin, Germany
| | - Kevin Pagel
- Fritz-Haber-Institut der Max-Planck-Gesellschaft, Faradayweg 4-6, 14195 Berlin, Germany.,Institut für Chemie und Biochemie, Freie Universität Berlin, Arnimallee 22, 14195 Berlin, Germany
| |
Collapse
|
12
|
Macrophages bind LDL using heparan sulfate and the perlecan protein core. J Biol Chem 2021; 296:100520. [PMID: 33684447 PMCID: PMC8027565 DOI: 10.1016/j.jbc.2021.100520] [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] [Received: 12/10/2020] [Revised: 03/03/2021] [Accepted: 03/04/2021] [Indexed: 12/25/2022] Open
Abstract
The retention of low-density lipoprotein (LDL) is a key process in the pathogenesis of atherosclerosis and largely mediated via smooth-muscle cell-derived extracellular proteoglycans including the glycosaminoglycan chains. Macrophages can also internalize lipids via complexes with proteoglycans. However, the role of polarized macrophage-derived proteoglycans in binding LDL is unknown and important to advance our understanding of the pathogenesis of atherosclerosis. We therefore examined the identity of proteoglycans, including the pendent glycosaminoglycans, produced by polarized macrophages to gain insight into the molecular basis for LDL binding. Using the quartz crystal microbalance with dissipation monitoring technique, we established that classically activated macrophage (M1)- and alternatively activated macrophage (M2)-derived proteoglycans bind LDL via both the protein core and heparan sulfate (HS) in vitro. Among the proteoglycans secreted by macrophages, we found perlecan was the major protein core that bound LDL. In addition, we identified perlecan in the necrotic core as well as the fibrous cap of advanced human atherosclerotic lesions in the same regions as HS and colocalized with M2 macrophages, suggesting a functional role in lipid retention in vivo. These findings suggest that macrophages may contribute to LDL retention in the plaque by the production of proteoglycans; however, their contribution likely depends on both their phenotype within the plaque and the presence of enzymes, such as heparanase, that alter the secreted protein structure.
Collapse
|
13
|
Aregueta-Robles UA, Enke YL, Carter PM, Green RA, Poole-Warren LA. Subthreshold Electrical Stimulation for Controlling Protein-Mediated Impedance Increases in Platinum Cochlear Electrode. IEEE Trans Biomed Eng 2020; 67:3510-3520. [DOI: 10.1109/tbme.2020.2989754] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
|
14
|
Lord MS, Melrose J, Day AJ, Whitelock JM. The Inter-α-Trypsin Inhibitor Family: Versatile Molecules in Biology and Pathology. J Histochem Cytochem 2020; 68:907-927. [PMID: 32639183 DOI: 10.1369/0022155420940067] [Citation(s) in RCA: 78] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Inter-α-trypsin inhibitor (IαI) family members are ancient and unique molecules that have evolved over several hundred million years of vertebrate evolution. IαI is a complex containing the proteoglycan bikunin to which heavy chain proteins are covalently attached to the chondroitin sulfate chain. Besides its matrix protective activity through protease inhibitory action, IαI family members interact with extracellular matrix molecules and most notably hyaluronan, inhibit complement, and provide cell regulatory functions. Recent evidence for the diverse roles of the IαI family in both biology and pathology is reviewed and gives insight into their pivotal roles in tissue homeostasis. In addition, the clinical uses of these molecules are explored, such as in the treatment of inflammatory conditions including sepsis and Kawasaki disease, which has recently been associated with severe acute respiratory syndrome coronavirus 2 infection in children.
Collapse
Affiliation(s)
- Megan S Lord
- Graduate School of Biomedical Engineering, UNSW Sydney, Sydney, NSW, Australia
| | - James Melrose
- Graduate School of Biomedical Engineering, UNSW Sydney, Sydney, NSW, Australia.,Raymond Purves Bone and Joint Research Laboratories, Kolling Institute of Medical Research, Royal North Shore Hospital and University of Sydney, St. Leonards, NSW, Australia.,Sydney Medical School, Northern, Sydney University, Royal North Shore Hospital, St. Leonards, NSW, Australia
| | - Anthony J Day
- Wellcome Trust Centre for Cell-Matrix Research and Lydia Becker Institute of Immunology and Inflammation, Division of Cell-Matrix Biology and Regenerative Medicine, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester, UK
| | - John M Whitelock
- Graduate School of Biomedical Engineering, UNSW Sydney, Sydney, NSW, Australia.,Stem Cell Extracellular Matrix & Glycobiology, Wolfson Centre for Stem Cells, Tissue Engineering and Modelling, Faculty of Medicine, University of Nottingham, Nottingham, UK
| |
Collapse
|
15
|
Ramadan S, Li T, Yang W, Zhang J, Rashidijahanabad Z, Tan Z, Parameswaran N, Huang X. Chemical Synthesis and Anti-Inflammatory Activity of Bikunin Associated Chondroitin Sulfate 24-mer. ACS CENTRAL SCIENCE 2020; 6:913-920. [PMID: 32607438 PMCID: PMC7318065 DOI: 10.1021/acscentsci.9b01199] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/19/2019] [Indexed: 05/09/2023]
Abstract
Bikunin, a chondroitin sulfate (CS) proteoglycan clinically used to treat acute inflammation and sepsis, contains a CS chain with more than 20 monosaccharide units. To understand the function of the CS chain of bikunin, synthesis of long CS chains is needed. After exploring multiple glycosylation approaches and protective group chemistry, we report herein the successful generation of the longest CS chain to date (24-mer) in an excellent overall yield on a multi-mg scale. The anti-inflammatory activities of both bikunin and the synthetic 24-mer were determined, and the results demonstrate that both the glycan and the core protein are important for anti-inflammatory activities of bikunin by reducing macrophage production of proinflammatory cytokines.
Collapse
Affiliation(s)
- Sherif Ramadan
- Department
of Chemistry, Michigan State University, 578 South Shaw Lane, East Lansing, Michigan 48824, United States
- Chemistry
Department, Faculty of Science, Benha University, Benha, Qaliobiya 13518, Egypt
- Institute
for Quantitative Health Science and Engineering, Michigan State University, East Lansing, Michigan 48824, United States
| | - Tianlu Li
- Department
of Chemistry, Michigan State University, 578 South Shaw Lane, East Lansing, Michigan 48824, United States
- Institute
for Quantitative Health Science and Engineering, Michigan State University, East Lansing, Michigan 48824, United States
| | - Weizhun Yang
- Department
of Chemistry, Michigan State University, 578 South Shaw Lane, East Lansing, Michigan 48824, United States
- Institute
for Quantitative Health Science and Engineering, Michigan State University, East Lansing, Michigan 48824, United States
| | - Jicheng Zhang
- Department
of Chemistry, Michigan State University, 578 South Shaw Lane, East Lansing, Michigan 48824, United States
- Institute
for Quantitative Health Science and Engineering, Michigan State University, East Lansing, Michigan 48824, United States
| | - Zahra Rashidijahanabad
- Department
of Chemistry, Michigan State University, 578 South Shaw Lane, East Lansing, Michigan 48824, United States
- Institute
for Quantitative Health Science and Engineering, Michigan State University, East Lansing, Michigan 48824, United States
| | - Zibin Tan
- Department
of Chemistry, Michigan State University, 578 South Shaw Lane, East Lansing, Michigan 48824, United States
- Institute
for Quantitative Health Science and Engineering, Michigan State University, East Lansing, Michigan 48824, United States
| | - Narayanan Parameswaran
- Department
of Physiology, Michigan State University, East Lansing, Michigan 48824, United States
| | - Xuefei Huang
- Department
of Chemistry, Michigan State University, 578 South Shaw Lane, East Lansing, Michigan 48824, United States
- Institute
for Quantitative Health Science and Engineering, Michigan State University, East Lansing, Michigan 48824, United States
- Department
of Biomedical Engineering, Michigan State
University, East Lansing, Michigan 48824, United States
- E-mail:
| |
Collapse
|
16
|
Lepedda AJ, De Muro P, Capobianco G, Formato M. Role of the small proteoglycan bikunin in human reproduction. Hormones (Athens) 2020; 19:123-133. [PMID: 31728877 DOI: 10.1007/s42000-019-00149-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/12/2019] [Accepted: 10/21/2019] [Indexed: 02/06/2023]
Abstract
PURPOSE Female reproductive events, including ovulation, menstruation, implantation, and delivery, are physiologically characterized by deep tissue remodeling and display hallmark signs of inflammation. This review discusses the pleiotropic roles played by bikunin in human reproduction. METHODS A comprehensive literature search of the Medline/PubMed database was performed on the following topics: bikunin structure, roles in pathophysiological conditions and involvement in human reproduction, and usefulness as a marker of gestational complications or as a drug to improve pregnancy outcomes. RESULTS Bikunin is a small chondroitin sulfate proteoglycan found in blood, urine, and amniotic and cerebrospinal fluids, known for its anti-inflammatory and anti-proteolytic activities. Its levels are usually low, but they can increase several-fold in both acute and chronic inflammatory diseases. Bikunin plays key roles in reproductive events, such as cumulus-oocyte complex formation, pregnancy, and delivery. Its levels have been associated with the most common pregnancy complications such as preterm delivery, pre-eclampsia, and gestational diabetes mellitus. Finally, its intravaginal administration has been reported to reduce the risk of preterm delivery and to improve neonatal outcomes. CONCLUSIONS Because of its pleiotropic roles in several reproductive events and its association with some life-threatening pathological conditions of pregnancy, bikunin may represent a non-invasive marker for improving follow-up and early diagnosis. Studies showing its usefulness as a drug for reducing the risk of preterm delivery and improving neonatal outcomes have yielded interesting results that deserve to be investigated through further research.
Collapse
Affiliation(s)
- Antonio Junior Lepedda
- Department of Biomedical Sciences, University of Sassari, Viale San Pietro 43, 07100, Sassari, Italy
| | - Pierina De Muro
- Department of Biomedical Sciences, University of Sassari, Viale San Pietro 43, 07100, Sassari, Italy
| | - Giampiero Capobianco
- Gynecologic and Obstetric Clinic, Department of Medical, Surgical and Experimental Sciences, University of Sassari, Viale San Pietro 12, 07100, Sassari, Italy.
| | - Marilena Formato
- Department of Biomedical Sciences, University of Sassari, Viale San Pietro 43, 07100, Sassari, Italy.
| |
Collapse
|
17
|
Pomin VH, Vignovich WP, Gonzales AV, Vasconcelos AA, Mulloy B. Galactosaminoglycans: Medical Applications and Drawbacks. Molecules 2019; 24:E2803. [PMID: 31374852 PMCID: PMC6696379 DOI: 10.3390/molecules24152803] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2019] [Revised: 07/24/2019] [Accepted: 07/30/2019] [Indexed: 12/28/2022] Open
Abstract
Galactosaminoglycans (GalAGs) are sulfated glycans composed of alternating N-acetylgalactosamine and uronic acid units. Uronic acid epimerization, sulfation patterns and fucosylation are modifications observed on these molecules. GalAGs have been extensively studied and exploited because of their multiple biomedical functions. Chondroitin sulfates (CSs), the main representative family of GalAGs, have been used in alternative therapy of joint pain/inflammation and osteoarthritis. The relatively novel fucosylated chondroitin sulfate (FCS), commonly found in sea cucumbers, has been screened in multiple systems in addition to its widely studied anticoagulant action. Biomedical properties of GalAGs are directly dependent on the sugar composition, presence or lack of fucose branches, as well as sulfation patterns. Although research interest in GalAGs has increased considerably over the three last decades, perhaps motivated by the parallel progress of glycomics, serious questions concerning the effectiveness and potential side effects of GalAGs have recently been raised. Doubts have centered particularly on the beneficial functions of CS-based therapeutic supplements and the potential harmful effects of FCS as similarly observed for oversulfated chondroitin sulfate, as a contaminant of heparin. Unexpected components were also detected in CS-based pharmaceutical preparations. This review therefore aims to offer a discussion on (1) the current and potential therapeutic applications of GalAGs, including those of unique features extracted from marine sources, and (2) the potential drawbacks of this class of molecules when applied to medicine.
Collapse
Affiliation(s)
- Vitor H Pomin
- Department of Biomolecular Sciences, School of Pharmacy, University of Mississippi, Oxford, MS 38677-1848, USA.
- Research Institute of Pharmaceutical Sciences, School of Pharmacy, University of Mississippi, Oxford, MS 38677-1848, USA.
| | - William P Vignovich
- Department of Biomolecular Sciences, School of Pharmacy, University of Mississippi, Oxford, MS 38677-1848, USA
| | - Alysia V Gonzales
- Department of Biomolecular Sciences, School of Pharmacy, University of Mississippi, Oxford, MS 38677-1848, USA
| | - Ariana A Vasconcelos
- Institute of Medical Biochemistry Leopoldo de Meis, Federal University of Rio de Janeiro, Rio de Janeiro, RJ 21941-590, Brazil
| | - Barbara Mulloy
- Imperial College, Department of Medicine, Burlington Danes Building, Du Cane Road, London W12 0NN, UK
| |
Collapse
|
18
|
Hayes AJ, Melrose J. Glycosaminoglycan and Proteoglycan Biotherapeutics in Articular Cartilage Protection and Repair Strategies: Novel Approaches to Visco‐supplementation in Orthobiologics. ADVANCED THERAPEUTICS 2019. [DOI: 10.1002/adtp.201900034] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Affiliation(s)
- Anthony J. Hayes
- Bioimaging Research HubCardiff School of BiosciencesCardiff University Cardiff CF10 3AX Wales UK
| | - James Melrose
- Graduate School of Biomedical EngineeringUNSW Sydney Sydney NSW 2052 Australia
- Raymond Purves Bone and Joint Research LaboratoriesKolling Institute of Medical ResearchRoyal North Shore Hospital and The Faculty of Medicine and HealthUniversity of Sydney St. Leonards NSW 2065 Australia
- Sydney Medical SchoolNorthernRoyal North Shore HospitalSydney University St. Leonards NSW 2065 Australia
| |
Collapse
|
19
|
Hascall VC. The journey of hyaluronan research in the Journal of Biological Chemistry. J Biol Chem 2019; 294:1690-1696. [PMID: 30710015 DOI: 10.1074/jbc.tm118.005836] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Hyaluronan has a very simple structure. It is a linear glycosaminoglycan composed of disaccharide units of GlcNAc and d-glucuronic acid with alternating β-1,4 and β-1,3 glycosidic bonds that can be repeated 20,000 or more times, a molecular mass >8 million Da, and a length >20 μm. However, it has a very complex biology. It is a major, ubiquitous component of extracellular matrices involved in everything from fertilization, development, inflammations, to cancer. This JBC Review highlights some of these processes that were initiated through publications in the Journal of Biological Chemistry.
Collapse
Affiliation(s)
- Vincent C Hascall
- Department of Biomedical Engineering, Cleveland Clinic, Cleveland, Ohio 44195.
| |
Collapse
|
20
|
Smith SM, Melrose J. A Retrospective Analysis of the Cartilage Kunitz Protease Inhibitory Proteins Identifies These as Members of the Inter-α-Trypsin Inhibitor Superfamily with Potential Roles in the Protection of the Articulatory Surface. Int J Mol Sci 2019; 20:ijms20030497. [PMID: 30678366 PMCID: PMC6387120 DOI: 10.3390/ijms20030497] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2018] [Revised: 01/17/2019] [Accepted: 01/21/2019] [Indexed: 02/06/2023] Open
Abstract
Aim: The aim of this study was to assess if the ovine articular cartilage serine proteinase inhibitors (SPIs) were related to the Kunitz inter-α-trypsin inhibitor (ITI) family. Methods: Ovine articular cartilage was finely diced and extracted in 6 M urea and SPIs isolated by sequential anion exchange, HA affinity and Sephadex G100 gel permeation chromatography. Selected samples were also subjected to chymotrypsin and concanavalin-A affinity chromatography. Eluant fractions from these isolation steps were monitored for protein and trypsin inhibitory activity. Inhibitory fractions were assessed by affinity blotting using biotinylated trypsin to detect SPIs and by Western blotting using antibodies to α1-microglobulin, bikunin, TSG-6 and 2-B-6 (+) CS epitope generated by chondroitinase-ABC digestion. Results: 2-B-6 (+) positive 250, 220,120, 58 and 36 kDa SPIs were detected. The 58 kDa SPI contained α1-microglobulin, bikunin and chondroitin-4-sulfate stub epitope consistent with an identity of α1-microglobulin-bikunin (AMBP) precursor and was also isolated by concanavalin-A lectin affinity chromatography indicating it had N-glycosylation. Kunitz protease inhibitor (KPI) species of 36, 26, 12 and 6 kDa were autolytically generated by prolonged storage of the 120 and 58 kDa SPIs; chymotrypsin affinity chromatography generated the 6 kDa SPI. KPI domain 1 and 2 SPIs were separated by concanavalin lectin affinity chromatography, domain 1 displayed affinity for this lectin indicating it had N-glycosylation. KPI 1 and 2 displayed potent inhibitory activity against trypsin, chymotrypsin, kallikrein, leucocyte elastase and cathepsin G. Localisation of versican, lubricin and hyaluronan (HA) in the surface regions of articular cartilage represented probable binding sites for the ITI serine proteinase inhibitors (SPIs) which may preserve articulatory properties and joint function. Discussion/Conclusions: The Kunitz SPI proteins synthesised by articular chondrocytes are members of the ITI superfamily. By analogy with other tissues in which these proteins occur we deduce that the cartilage Kunitz SPIs may be multifunctional proteins. Binding of the cartilage Kunitz SPIs to HA may protect this polymer from depolymerisation by free radical damage and may also protect other components in the cartilage surface from proteolytic degradation preserving joint function.
Collapse
Affiliation(s)
- Susan M Smith
- Raymond Purves Bone and Joint Research Laboratory, Kolling Institute, Northern Sydney Local Health District, St. Leonards, NSW 2065, Australia.
| | - James Melrose
- Raymond Purves Bone and Joint Research Laboratory, Kolling Institute, Northern Sydney Local Health District, St. Leonards, NSW 2065, Australia.
- Graduate School of Biomedical Engineering, University of New South Wales, Sydney, NSW 2052, Australia.
- Sydney Medical School, Northern, The University of Sydney, Royal North Shore Hospital, St. Leonards, NSW 2065, Australia.
- Faculty of Medicine and Health, University of Sydney, Royal North Shore Hospital, St. Leonards, NSW 2065, Australia.
| |
Collapse
|
21
|
Palmer JC, Green RA, Boscher F, Poole-Warren LA, Carter PM, Enke YL, Lovell NH, Lord MS. Development and performance of a biomimetic artificial perilymph for in vitro testing of medical devices. J Neural Eng 2019; 16:026006. [DOI: 10.1088/1741-2552/aaf482] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
|
22
|
Biodiversity of CS–proteoglycan sulphation motifs: chemical messenger recognition modules with roles in information transfer, control of cellular behaviour and tissue morphogenesis. Biochem J 2018; 475:587-620. [DOI: 10.1042/bcj20170820] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2017] [Revised: 12/20/2017] [Accepted: 01/07/2018] [Indexed: 12/19/2022]
Abstract
Chondroitin sulphate (CS) glycosaminoglycan chains on cell and extracellular matrix proteoglycans (PGs) can no longer be regarded as merely hydrodynamic space fillers. Overwhelming evidence over recent years indicates that sulphation motif sequences within the CS chain structure are a source of significant biological information to cells and their surrounding environment. CS sulphation motifs have been shown to interact with a wide variety of bioactive molecules, e.g. cytokines, growth factors, chemokines, morphogenetic proteins, enzymes and enzyme inhibitors, as well as structural components within the extracellular milieu. They are therefore capable of modulating a panoply of signalling pathways, thus controlling diverse cellular behaviours including proliferation, differentiation, migration and matrix synthesis. Consequently, through these motifs, CS PGs play significant roles in the maintenance of tissue homeostasis, morphogenesis, development, growth and disease. Here, we review (i) the biodiversity of CS PGs and their sulphation motif sequences and (ii) the current understanding of the signalling roles they play in regulating cellular behaviour during tissue development, growth, disease and repair.
Collapse
|
23
|
Day AJ, Milner CM. TSG-6: A multifunctional protein with anti-inflammatory and tissue-protective properties. Matrix Biol 2018; 78-79:60-83. [PMID: 29362135 DOI: 10.1016/j.matbio.2018.01.011] [Citation(s) in RCA: 206] [Impact Index Per Article: 29.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2017] [Revised: 01/09/2018] [Accepted: 01/11/2018] [Indexed: 02/06/2023]
Abstract
Tumor necrosis factor- (TNF) stimulated gene-6 (TSG-6) is an inflammation-associated secreted protein that has been implicated as having important and diverse tissue protective and anti-inflammatory properties, e.g. mediating many of the immunomodulatory and beneficial activities of mesenchymal stem/stromal cells. TSG-6 is constitutively expressed in some tissues, which are either highly metabolically active or subject to challenges from the environment, perhaps providing protection in these contexts. The diversity of its functions are dependent on the binding of TSG-6 to numerous ligands, including matrix molecules such as glycosaminoglycans, as well as immune regulators and growth factors that themselves interact with these linear polysaccharides. It is becoming apparent that TSG-6 can directly affect matrix structure and modulate the way extracellular signalling molecules interact with matrix. In this review, we focus mainly on the literature for TSG-6 over the last 10 years, summarizing its expression, structure, ligand-binding properties, biological functions and highlighting TSG-6's potential as a therapeutic for a broad range of disease indications.
Collapse
Affiliation(s)
- Anthony J Day
- Wellcome Trust Centre for Cell-Matrix Research, Division of Cell-Matrix Biology and Regenerative Medicine, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, Michael Smith Building, Oxford Road, Manchester M13 9PT, UK.
| | - Caroline M Milner
- Division of Cell-Matrix Biology and Regenerative Medicine, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, Michael Smith Building, Oxford Road, Manchester M13 9PT, UK.
| |
Collapse
|
24
|
Rnjak-Kovacina J, Tang F, Lin X, Whitelock JM, Lord MS. Recombinant Domain V of Human Perlecan Is a Bioactive Vascular Proteoglycan. Biotechnol J 2017; 12. [PMID: 28846206 DOI: 10.1002/biot.201700196] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2017] [Revised: 07/17/2017] [Indexed: 11/12/2022]
Abstract
The C-terminal domain V of the extracellular matrix proteoglycan perlecan plays unique and often divergent roles in a number of biological processes, including angiogenesis, vascular cell interactions, wound healing, and autophagy. Recombinant forms of domain V have been proposed as therapeutic agents for the treatment of cancer, stroke, and the development of cardiovascular devices and bioartificial tissues. However, the effect of domain V appears to be related to the differences in domain V structure and function observed in different expression systems and environments and exactly how this occurs is not well understood. In this study, the sequence from amino acid 3626 to 4391 of the perlecan protein core, which includes domain V, is expressed in HEK-293 cells and purified as a secreted product from conditioned media. This recombinant domain V (rDV) is expressed as a proteoglycan decorated with heparan sulfate and chondroitin sulfate chains and supports endothelial cell interactions to the same extent as full-length perlecan. This expression system serves as an important model of recombinant proteoglycan expression, as well as a source of biologically active rDV for therapeutic applications.
Collapse
Affiliation(s)
| | - Fengying Tang
- Graduate School of Biomedical Engineering, UNSW Sydney, Sydney, Australia
| | - Xiaoting Lin
- Graduate School of Biomedical Engineering, UNSW Sydney, Sydney, Australia
| | - John M Whitelock
- Graduate School of Biomedical Engineering, UNSW Sydney, Sydney, Australia
| | - Megan S Lord
- Graduate School of Biomedical Engineering, UNSW Sydney, Sydney, Australia
| |
Collapse
|
25
|
Nilsson J, Noborn F, Gomez Toledo A, Nasir W, Sihlbom C, Larson G. Characterization of Glycan Structures of Chondroitin Sulfate-Glycopeptides Facilitated by Sodium Ion-Pairing and Positive Mode LC-MS/MS. JOURNAL OF THE AMERICAN SOCIETY FOR MASS SPECTROMETRY 2017; 28:229-241. [PMID: 27873218 PMCID: PMC5227003 DOI: 10.1007/s13361-016-1539-1] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/01/2016] [Revised: 10/13/2016] [Accepted: 10/20/2016] [Indexed: 06/06/2023]
Abstract
Purification and liquid chromatography-tandem mass spectrometry (LC-MS/MS) characterization of glycopeptides, originating from protease digests of glycoproteins, enables site-specific analysis of protein N- and O-glycosylations. We have described a protocol to enrich, hydrolyze by chondroitinase ABC, and characterize chondroitin sulfate-containing glycopeptides (CS-glycopeptides) using positive mode LC-MS/MS. The CS-glycopeptides, originating from the Bikunin proteoglycan of human urine samples, had ΔHexAGalNAcGlcAGalGalXyl-O-Ser hexasaccharide structure and were further substituted with 0-3 sulfate and 0-1 phosphate groups. However, it was not possible to exactly pinpoint sulfate attachment residues, for protonated precursors, due to extensive fragmentation of sulfate groups using high-energy collision induced dissociation (HCD). To circumvent the well-recognized sulfate instability, we now introduced Na+ ions to form sodiated precursors, which protected sulfate groups from decomposition and facilitated the assignment of sulfate modifications. Sulfate groups were pinpointed to both Gal residues and to the GalNAc of the hexasaccharide structure. The intensities of protonated and sodiated saccharide oxonium ions were very prominent in the HCD-MS2 spectra, which provided complementary structural analysis of sulfate substituents of CS-glycopeptides. We have demonstrated a considerable heterogeneity of the bikunin CS linkage region. The realization of these structural variants should be beneficial in studies aimed at investigating the importance of the CS linkage region with regards to the biosynthesis of CS and potential interactions to CS binding proteins. Also, the combined use of protonated and sodiated precursors for positive mode HCD fragmentation analysis will likely become useful for additional classes of sulfated glycopeptides. Graphical Abstract ᅟ.
Collapse
Affiliation(s)
- Jonas Nilsson
- Department of Clinical Chemistry and Transfusion Medicine, Institute of Biomedicine, Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - Fredrik Noborn
- Department of Clinical Chemistry and Transfusion Medicine, Institute of Biomedicine, Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - Alejandro Gomez Toledo
- Department of Clinical Chemistry and Transfusion Medicine, Institute of Biomedicine, Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - Waqas Nasir
- Department of Clinical Chemistry and Transfusion Medicine, Institute of Biomedicine, Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - Carina Sihlbom
- The Proteomics Core Facility, Core Facilities, Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - Göran Larson
- Department of Clinical Chemistry and Transfusion Medicine, Institute of Biomedicine, Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden.
| |
Collapse
|
26
|
Park Y, Jowitt TA, Day AJ, Prestegard JH. Nuclear Magnetic Resonance Insight into the Multiple Glycosaminoglycan Binding Modes of the Link Module from Human TSG-6. Biochemistry 2016; 55:262-76. [PMID: 26685054 PMCID: PMC5073374 DOI: 10.1021/acs.biochem.5b01148] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Tumor necrosis factor-stimulated gene-6 (TSG-6) is a hyaluronan (HA)-binding protein that is essential for stabilizing and remodeling the extracellular matrix (ECM) during ovulation and inflammatory disease processes such as arthritis. The Link module, one of the domains of TSG-6, is responsible for binding hyaluronan and other glycosaminoglycans found in the ECM. In this study, we used a well-defined chondroitin sulfate (CS) hexasaccharide (ΔC444S) to determine the structure of the Link module, in solution, in its chondroitin sulfate-bound state. A variety of nuclear magnetic resonance techniques were employed, including chemical shift perturbation, residual dipolar couplings (RDCs), nuclear Overhauser effects, spin relaxation measurements, and paramagnetic relaxation enhancements from a spin-labeled analogue of ΔC444S. The binding site for ΔC444S on the Link module overlapped with that of HA. Surprisingly, ΔC444S binding induced dimerization of the Link module (as confirmed by analytical ultracentrifugation), and a second weak binding site that partially overlapped with a previously identified heparin site was detected. A dimer model was generated using chemical shift perturbations and RDCs as restraints in the docking program HADDOCK. We postulate that the molecular cross-linking enhanced by the multiple binding modes of the Link module might be critical for remodeling the ECM during inflammation/ovulation and might contribute to other functions of TSG-6.
Collapse
Affiliation(s)
- Younghee Park
- Complex Carbohydrate Research Center, 315 Riverbend Road, University of Georgia, Athens, GA 30602, USA
| | - Thomas A. Jowitt
- Wellcome Trust Centre for Cell-Matrix Research, Faculty of Life Sciences, University of Manchester, Oxford Road, Manchester M13 9PT, UK
| | - Anthony J. Day
- Wellcome Trust Centre for Cell-Matrix Research, Faculty of Life Sciences, University of Manchester, Oxford Road, Manchester M13 9PT, UK
| | - James H. Prestegard
- Complex Carbohydrate Research Center, 315 Riverbend Road, University of Georgia, Athens, GA 30602, USA
| |
Collapse
|
27
|
Briggs DC, Birchenough HL, Ali T, Rugg MS, Waltho JP, Ievoli E, Jowitt TA, Enghild JJ, Richter RP, Salustri A, Milner CM, Day AJ. Metal Ion-dependent Heavy Chain Transfer Activity of TSG-6 Mediates Assembly of the Cumulus-Oocyte Matrix. J Biol Chem 2015; 290:28708-23. [PMID: 26468290 PMCID: PMC4661386 DOI: 10.1074/jbc.m115.669838] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2015] [Indexed: 11/06/2022] Open
Abstract
The matrix polysaccharide hyaluronan (HA) has a critical role in the expansion of the cumulus cell-oocyte complex (COC), a process that is necessary for ovulation and fertilization in most mammals. Hyaluronan is organized into a cross-linked network by the cooperative action of three proteins, inter-α-inhibitor (IαI), pentraxin-3, and TNF-stimulated gene-6 (TSG-6), driving the expansion of the COC and providing the cumulus matrix with its required viscoelastic properties. Although it is known that matrix stabilization involves the TSG-6-mediated transfer of IαI heavy chains (HCs) onto hyaluronan (to form covalent HC·HA complexes that are cross-linked by pentraxin-3) and that this occurs via the formation of covalent HC·TSG-6 intermediates, the underlying molecular mechanisms are not well understood. Here, we have determined the tertiary structure of the CUB module from human TSG-6, identifying a calcium ion-binding site and chelating glutamic acid residue that mediate the formation of HC·TSG-6. This occurs via an initial metal ion-dependent, non-covalent, interaction between TSG-6 and HCs that also requires the presence of an HC-associated magnesium ion. In addition, we have found that the well characterized hyaluronan-binding site in the TSG-6 Link module is not used for recognition during transfer of HCs onto HA. Analysis of TSG-6 mutants (with impaired transferase and/or hyaluronan-binding functions) revealed that although the TSG-6-mediated formation of HC·HA complexes is essential for the expansion of mouse COCs in vitro, the hyaluronan-binding function of TSG-6 does not play a major role in the stabilization of the murine cumulus matrix.
Collapse
Affiliation(s)
- David C Briggs
- From the Wellcome Trust Centre for Cell-Matrix Research and the Faculty of Life Sciences, University of Manchester, Oxford Road, Manchester M13 9PT, United Kingdom
| | - Holly L Birchenough
- From the Wellcome Trust Centre for Cell-Matrix Research and the Faculty of Life Sciences, University of Manchester, Oxford Road, Manchester M13 9PT, United Kingdom
| | - Tariq Ali
- From the Wellcome Trust Centre for Cell-Matrix Research and the Faculty of Life Sciences, University of Manchester, Oxford Road, Manchester M13 9PT, United Kingdom
| | - Marilyn S Rugg
- the Medical Research Council Immunochemistry Unit, Department of Biochemistry, University of Oxford, South Parks Road, Oxford OX1 3QU, United Kingdom
| | - Jon P Waltho
- the Faculty of Life Sciences, University of Manchester, Oxford Road, Manchester M13 9PT, United Kingdom
| | - Elena Ievoli
- the Department of Biomedicine and Prevention, University of Rome Tor Vergata, Rome 00133, Italy
| | - Thomas A Jowitt
- From the Wellcome Trust Centre for Cell-Matrix Research and the Faculty of Life Sciences, University of Manchester, Oxford Road, Manchester M13 9PT, United Kingdom
| | - Jan J Enghild
- the Department of Molecular Chemistry, University of Aarhus, 8000 Aarhus C, Denmark
| | - Ralf P Richter
- CIC biomaGUNE, 20009 Donostia-San Sebastian, Spain, the Department of Molecular Chemistry, University Grenoble Alpes and CNRS, 38000 Grenoble, France, and the Max Planck Institute for Intelligent Systems, 70569 Stuttgart, Germany
| | - Antonietta Salustri
- the Department of Biomedicine and Prevention, University of Rome Tor Vergata, Rome 00133, Italy
| | - Caroline M Milner
- the Faculty of Life Sciences, University of Manchester, Oxford Road, Manchester M13 9PT, United Kingdom
| | - Anthony J Day
- From the Wellcome Trust Centre for Cell-Matrix Research and the Faculty of Life Sciences, University of Manchester, Oxford Road, Manchester M13 9PT, United Kingdom,
| |
Collapse
|
28
|
Sun X, Li L, Overdier KH, Ammons LA, Douglas IS, Burlew CC, Zhang F, Schmidt EP, Chi L, Linhardt RJ. Analysis of Total Human Urinary Glycosaminoglycan Disaccharides by Liquid Chromatography-Tandem Mass Spectrometry. Anal Chem 2015; 87:6220-7. [PMID: 26005898 PMCID: PMC4822829 DOI: 10.1021/acs.analchem.5b00913] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
The determination of complex analytes, present at low concentrations, in biological fluids poses a difficult challenge. This study relies on an optimized method of recovery, enzymatic treatment, and disaccharide analysis by liquid chromatography-tandem mass spectrometry to rapidly determine low concentrations of glycosaminoglycans in human urine. The approach utilizes multiple reaction monitoring (MRM) of glycosaminoglycan disaccharides obtained from treating urine samples with recombinant heparin lyases and chondroitin lyase. This rapid and sensitive method allows the analysis of glycosaminoglycan content and disaccharide composition in urine samples having concentrations 10- to 100-fold lower than those typically analyzed from patients with metabolic diseases, such as mucopolysaccharidosis. The current method facilitates the analysis low (ng/mL) levels of urinary glycosaminoglycans present in healthy individuals and in patients with pathological conditions, such as inflammation and cancers, that can subtly alter glycosaminoglycan content and composition.
Collapse
Affiliation(s)
- Xiaojun Sun
- National Glycoengineering Research Center, Shandong University, Jinan 250100, China
- Department of Chemistry and Chemical Biology, Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, New York 12180, United States
| | - Lingyun Li
- Department of Chemistry and Chemical Biology, Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, New York 12180, United States
- Wadsworth Center, New York State Department of Health, Albany, New York 12201, United States
| | - Katherine H. Overdier
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, Denver, Colorado 80204, United States
| | - Lee Anne Ammons
- Department of Surgery, Denver Health Medical Center, Denver, Colorado 80204, United States
| | - Ivor S. Douglas
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, Denver, Colorado 80204, United States
| | - Clay Cothren Burlew
- Department of Surgery, Denver Health Medical Center, Denver, Colorado 80204, United States
| | - Fuming Zhang
- Department of Chemical and Biological Engineering, Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, New York 12180, United States
| | - Eric P. Schmidt
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, Denver, Colorado 80204, United States
- Program in Translational Lung Research, Department of Medicine, Division of Pulmonary Sciences and Critical Care Medicine, University of Colorado School of Medicine, Aurora, Colorado 80045, United States
| | - Lianli Chi
- National Glycoengineering Research Center, Shandong University, Jinan 250100, China
| | - Robert J. Linhardt
- Department of Chemistry and Chemical Biology, Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, New York 12180, United States
- Department of Chemical and Biological Engineering, Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, New York 12180, United States
- Department of Biology, Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, New York 12180, United States
- Department of Biomedical Engineering, Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, New York 12180, United States
| |
Collapse
|
29
|
Biology and biotechnology of hyaluronan. Glycoconj J 2015; 32:93-103. [PMID: 25971701 DOI: 10.1007/s10719-015-9586-6] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2015] [Revised: 03/13/2015] [Accepted: 03/25/2015] [Indexed: 10/23/2022]
Abstract
The hyaluronan (HA) polymer is a critical component of extracellular matrix with a remarkable structure: is a linear and unbranched polymer without sulphate or phosphate groups. It is ubiquitous in mammals showing several biological functions, ranging from cell proliferation and migration to angiogenesis and inflammation. For its critical biological functions the amount of HA in tissues is carefully controlled by different mechanisms including covalent modification of the synthetic enzymes and epigenetic control of their gene expression. The concentration of HA is also critical in several pathologies including cancer, diabetes and inflammation. Beside these biological roles, the structural properties of HA allow it to take advantage of its capacity to form gels even at concentration of 1 % producing scaffolds with very promising applications in regenerative medicine as biocompatible material for advanced therapeutic uses. In this review we highlight the biological aspects of HA addressing the mechanisms controlling the HA content in tissues as well as its role in important human pathologies. In the second part of the review we highlight the different use of HA polymers in the modern biotechnology.
Collapse
|
30
|
Lu X, Jiang K, Han L, Zhang M, Zhou Y, Ma Y, Zhou Y, Meng S. Sulfonation of curcuminoids: Characterization and contribution of individual SULT enzymes. Mol Nutr Food Res 2015; 59:634-45. [DOI: 10.1002/mnfr.201400493] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2014] [Revised: 12/25/2014] [Accepted: 12/30/2014] [Indexed: 01/01/2023]
Affiliation(s)
- Xiaoyue Lu
- Department of Pharmaceutics; School of Pharmacy; China Medical University; Shenyang Liaoning P. R. China
| | - Kunyu Jiang
- Department of Pharmaceutics; School of Pharmacy; China Medical University; Shenyang Liaoning P. R. China
| | - Long Han
- Department of Pharmaceutics; School of Pharmacy; China Medical University; Shenyang Liaoning P. R. China
| | - Maofan Zhang
- Department of Pharmaceutics; School of Pharmacy; China Medical University; Shenyang Liaoning P. R. China
| | - Yu Zhou
- Department of Pharmaceutics; School of Pharmacy; China Medical University; Shenyang Liaoning P. R. China
| | - Yinglin Ma
- Department of Pharmaceutics; School of Pharmacy; China Medical University; Shenyang Liaoning P. R. China
| | - Yiping Zhou
- Department of Pharmaceutics; School of Pharmacy; China Medical University; Shenyang Liaoning P. R. China
| | - Shengnan Meng
- Department of Pharmaceutics; School of Pharmacy; China Medical University; Shenyang Liaoning P. R. China
| |
Collapse
|
31
|
Inter-α inhibitor protein and its associated glycosaminoglycans protect against histone-induced injury. Blood 2015; 125:2286-96. [PMID: 25631771 DOI: 10.1182/blood-2014-06-582759] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2014] [Accepted: 01/20/2015] [Indexed: 11/20/2022] Open
Abstract
Extracellular histones are mediators of tissue injury and organ dysfunction; therefore they constitute potential therapeutic targets in sepsis, inflammation, and thrombosis. Histone cytotoxicity in vitro decreases in the presence of plasma. Here, we demonstrate that plasma inter-α inhibitor protein (IAIP) neutralizes the cytotoxic effects of histones and decreases histone-induced platelet aggregation. These effects are mediated through the negatively charged glycosaminoglycans (GAGs) chondroitin sulfate and high-molecular-weight hyaluronan (HMW-HA) associated with IAIP. Cell surface anionic glycosaminoglycans heparan sulfate and HA protect the cells against histone-mediated damage in vitro. Surface plasmon resonance showed that both IAIP and HMW-HA directly bind to recombinant histone H4. In vivo neutralization of histones with IAIP and HMW-HA prevented histone-induced thrombocytopenia, bleeding, and lung microvascular thrombosis, decreased neutrophil activation, and averted histone-induced production of inflammatory cytokines and chemokines. IAIP and HMW-HA colocalized with histones in necrotic tissues and areas that displayed neutrophil extracellular traps. Increasing amounts of IAIP-histone complexes detected in the plasma of septic baboons correlated with increase in histones and/or nucleosomes and consumption of plasma IAIP. Our data suggest that IAIP, chondroitin sulfate, and HMW-HA are potential therapeutic agents to protect against histone-induced cytotoxicity, coagulopathy, systemic inflammation, and organ damage during inflammatory conditions such as sepsis and trauma.
Collapse
|
32
|
Lamkin E, Cheng G, Calabro A, Hascall VC, Joo EJ, Li L, Linhardt RJ, Lauer ME. Heavy chain transfer by tumor necrosis factor-stimulated gene 6 to the bikunin proteoglycan. J Biol Chem 2015; 290:5156-5166. [PMID: 25561734 DOI: 10.1074/jbc.m114.636258] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
We present data that hyaluronan (HA) polysaccharides, about 14-86 monosaccharides in length, are capable of accepting only a single heavy chain (HC) from inter-α-inhibitor via transfer by tumor necrosis factor-stimulated gene 6 (TSG-6) and that this transfer is irreversible. We propose that either the sulfate groups (or the sulfation pattern) at the reducing end of the chondroitin sulfate (CS) chain of bikunin, or the core protein itself, enables the bikunin proteoglycan (PG) to accept more than a single HC and permits TSG-6 to transfer these HCs from its relatively small CS chain to HA. To test these hypotheses, we investigated HC transfer to the intact CS chain of the bikunin PG, and to the free chain of bikunin. We observed that both the free CS chain and the intact bikunin PG were only able to accept a single HC from inter-α-inhibitor via transfer by TSG-6 and that HCs could be swapped from the bikunin PG and its free CS chain to HA. Furthermore, a significant portion of the bikunin PG was unable to accept a single heavy chain. We discuss explanations for these observations, including the intracellular assembly of inter-α-inhibitor. In summary, these data demonstrate that the sulfation of the CS chain of bikunin and/or its core protein promote HC transfer by TSG-6 to its relatively short CS chain, although they are insufficient to enable the CS chain of bikunin to accept more than one HC in the absence of other cofactors.
Collapse
Affiliation(s)
| | | | | | | | - Eun Ji Joo
- the Departments of Chemistry and Chemical Biology, Chemical and Biological Engineering, Biology, and Biomedical Engineering, Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, New York 12180
| | - Lingyun Li
- the Departments of Chemistry and Chemical Biology, Chemical and Biological Engineering, Biology, and Biomedical Engineering, Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, New York 12180
| | - Robert J Linhardt
- the Departments of Chemistry and Chemical Biology, Chemical and Biological Engineering, Biology, and Biomedical Engineering, Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, New York 12180
| | - Mark E Lauer
- From the Department of Biomedical Engineering and.
| |
Collapse
|
33
|
Lord MS, Farrugia BL, Rnjak-Kovacina J, Whitelock JM. Current serological possibilities for the diagnosis of arthritis with special focus on proteins and proteoglycans from the extracellular matrix. Expert Rev Mol Diagn 2015; 15:77-95. [PMID: 25382274 DOI: 10.1586/14737159.2015.979158] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
Abstract
This review discusses our current understanding of how the expression and turnover of components of the cartilage extracellular matrix (ECM) have been investigated, both as molecular markers of arthritis and as indicators of disease progression. The cartilage ECM proteome is well studied; it contains proteoglycans (aggrecan, perlecan and inter-α-trypsin inhibitor), collagens and glycoproteins (cartilage oligomeric matrix protein, fibronectin and lubricin) that provide the structural and functional changes in arthritis. However, the changes that occur in the carbohydrate structures, including glycosaminoglycans, with disease are less well studied. Investigations of the cartilage ECM proteome have revealed many potential biomarkers of arthritis. However, a clinical diagnostic or multiplex assay is yet to be realized due to issues with specificity to the pathology of arthritis. The future search for clinical biomarkers of arthritis is likely to involve both protein and carbohydrate markers of the ECM through the application of glycoproteomics.
Collapse
Affiliation(s)
- Megan S Lord
- Graduate School of Biomedical Engineering, The University of New South Wales, Sydney, NSW 2052, Australia
| | | | | | | |
Collapse
|
34
|
Noborn F, Gomez Toledo A, Sihlbom C, Lengqvist J, Fries E, Kjellén L, Nilsson J, Larson G. Identification of chondroitin sulfate linkage region glycopeptides reveals prohormones as a novel class of proteoglycans. Mol Cell Proteomics 2014; 14:41-9. [PMID: 25326458 DOI: 10.1074/mcp.m114.043703] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Vertebrates produce various chondroitin sulfate proteoglycans (CSPGs) that are important structural components of cartilage and other connective tissues. CSPGs also contribute to the regulation of more specialized processes such as neurogenesis and angiogenesis. Although many aspects of CSPGs have been studied extensively, little is known of where the CS chains are attached on the core proteins and so far, only a limited number of CSPGs have been identified. Obtaining global information on glycan structures and attachment sites would contribute to our understanding of the complex proteoglycan structures and may also assist in assigning CSPG specific functions. In the present work, we have developed a glycoproteomics approach that characterizes CS linkage regions, attachment sites, and identities of core proteins. CSPGs were enriched from human urine and cerebrospinal fluid samples by strong-anion-exchange chromatography, digested with chondroitinase ABC, a specific CS-lyase used to reduce the CS chain lengths and subsequently analyzed by nLC-MS/MS with a novel glycopeptide search algorithm. The protocol enabled the identification of 13 novel CSPGs, in addition to 13 previously established CSPGs, demonstrating that this approach can be routinely used to characterize CSPGs in complex human samples. Surprisingly, five of the identified CSPGs are traditionally defined as prohormones (cholecystokinin, chromogranin A, neuropeptide W, secretogranin-1, and secretogranin-3), typically stored and secreted from granules of endocrine cells. We hypothesized that the CS side chain may influence the assembly and structural organization of secretory granules and applied surface plasmon resonance spectroscopy to show that CS actually promotes the assembly of chromogranin A core proteins in vitro. This activity required mild acidic pH and suggests that the CS-side chains may also influence the self-assembly of chromogranin A in vivo giving a possible explanation to previous observations that chromogranin A has an inherent property to assemble in the acidic milieu of secretory granules.
Collapse
Affiliation(s)
- Fredrik Noborn
- From the ‡Department of Clinical Chemistry and Transfusion Medicine, Institute of Biomedicine, University of Gothenburg, Sahlgrenska University Hospital, SE-413 45 Gothenburg, Sweden
| | - Alejandro Gomez Toledo
- From the ‡Department of Clinical Chemistry and Transfusion Medicine, Institute of Biomedicine, University of Gothenburg, Sahlgrenska University Hospital, SE-413 45 Gothenburg, Sweden
| | - Carina Sihlbom
- §Proteomics Core Facility, Sahlgrenska Academy, University of Gothenburg, Box 413, SE-405 30, Sweden
| | - Johan Lengqvist
- §Proteomics Core Facility, Sahlgrenska Academy, University of Gothenburg, Box 413, SE-405 30, Sweden
| | - Erik Fries
- ¶Department of Medical Biochemistry and Microbiology, Uppsala University, SE-751 23 Uppsala, Sweden
| | - Lena Kjellén
- ¶Department of Medical Biochemistry and Microbiology, Uppsala University, SE-751 23 Uppsala, Sweden
| | - Jonas Nilsson
- From the ‡Department of Clinical Chemistry and Transfusion Medicine, Institute of Biomedicine, University of Gothenburg, Sahlgrenska University Hospital, SE-413 45 Gothenburg, Sweden
| | - Göran Larson
- From the ‡Department of Clinical Chemistry and Transfusion Medicine, Institute of Biomedicine, University of Gothenburg, Sahlgrenska University Hospital, SE-413 45 Gothenburg, Sweden;
| |
Collapse
|