1
|
Gyimesi M, Oikari LE, Yu C, Sutherland HG, Nyholt DR, Griffiths LR, Van Wijnen AJ, Okolicsanyi RK, Haupt LM. CpG methylation changes in human mesenchymal and neural stem cells in response to in vitro niche modifications. Biochimie 2024; 223:147-157. [PMID: 38640996 DOI: 10.1016/j.biochi.2024.04.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Revised: 04/15/2024] [Accepted: 04/16/2024] [Indexed: 04/21/2024]
Abstract
Stem cell therapies hold promise in addressing the burden of neurodegenerative diseases with human embryonic neural stem cells (hNSC-H9s) and bone marrow-derived human mesenchymal stem cells (hMSCs) as viable candidates. The induction of hMSC neurospheres (hMSC-IN) generate a more lineage-restricted common neural progenitor-like cell population, potentially tunable by heparan sulfate proteoglycans (HSPGs). We examined CpG (5 mC) site methylation patterns using Illumina Infinium 850 K EPIC arrays in hNSC-H9, hMSCs and hMSC-IN cultures with HSPG agonist heparin at early and late phases of growth. We identified key regulatory CpG sites in syndecans (SDC2; SDC4) that potentially regulate gene expression in monolayers. Unique hMSC-IN hypomethylation in glypicans (GPC3; GPC4) underscore their significance in neural lineages with Sulfatase 1 and 2 (SULF1 &2) CpG methylation changes potentially driving the neurogenic shift. hMSC-INs methylation levels at SULF1 CpG sites and SULF2:cg25401628 were more closely aligned with hNSC-H9 cells than with hMSCs. We further suggest SOX2 regulation governed by lncSOX2-Overall Transcript (lncSOX2-OT) methylation changes with preferential activation of ENO2 over other neuronal markers within hMSC-INs. Our findings illuminate epigenetic dynamics governing neural lineage commitment of hMSC-INs offering insights for targeted mechanisms for regenerative medicine and therapeutic strategies.
Collapse
Affiliation(s)
- Martina Gyimesi
- Stem Cell and Neurogenesis Group, Genomics Research Centre, Centre for Genomics and Personalised Health, School of Biomedical Sciences, Queensland University of Technology, Australia
| | - Lotta E Oikari
- Stem Cell and Neurogenesis Group, Genomics Research Centre, Centre for Genomics and Personalised Health, School of Biomedical Sciences, Queensland University of Technology, Australia
| | - Chieh Yu
- Stem Cell and Neurogenesis Group, Genomics Research Centre, Centre for Genomics and Personalised Health, School of Biomedical Sciences, Queensland University of Technology, Australia
| | - Heidi G Sutherland
- Stem Cell and Neurogenesis Group, Genomics Research Centre, Centre for Genomics and Personalised Health, School of Biomedical Sciences, Queensland University of Technology, Australia
| | - Dale R Nyholt
- Statistical and Genomic Epidemiology Laboratory, School of Biomedical Sciences, Faculty of Health and Centre for Genomics and Personalised Health, Queensland University of Technology, Brisbane, QLD, Australia
| | - Lyn R Griffiths
- Stem Cell and Neurogenesis Group, Genomics Research Centre, Centre for Genomics and Personalised Health, School of Biomedical Sciences, Queensland University of Technology, Australia
| | | | - Rachel K Okolicsanyi
- Stem Cell and Neurogenesis Group, Genomics Research Centre, Centre for Genomics and Personalised Health, School of Biomedical Sciences, Queensland University of Technology, Australia; Max Planck Queensland Centre for the Materials Science of Extracellular Matrices, Australia
| | - Larisa M Haupt
- Stem Cell and Neurogenesis Group, Genomics Research Centre, Centre for Genomics and Personalised Health, School of Biomedical Sciences, Queensland University of Technology, Australia; ARC Training Centre for Cell and Tissue Engineering Technologies, Queensland University of Technology (QUT), Australia; Max Planck Queensland Centre for the Materials Science of Extracellular Matrices, Australia; Centre for Biomedical Technologies, Queensland University of Technology (QUT), 60 Musk Ave., Kelvin Grove, QLD 4059, Australia.
| |
Collapse
|
2
|
Timm BM, Follmar JL, Porell RN, Glass K, Thacker BE, Glass CA, Godula K. Human extracellular sulfatases use a dual mechanism for regulation of growth factor interactions with heparan sulfate proteoglycans. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.11.22.568358. [PMID: 38045270 PMCID: PMC10690288 DOI: 10.1101/2023.11.22.568358] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/05/2023]
Abstract
Membrane-associated heparan sulfate (HS) proteoglycans (PGs) contribute to the regulation of extracellular cellular signaling cues, such as growth factors (GFs) and chemokines, essential for normal organismal functions and implicated in various pathophysiologies. PGs accomplish this by presenting high affinity binding sites for GFs and their receptors through highly sulfated regions of their HS polysaccharide chains. The composition of HS, and thus GF-binding specificity, are determined during biosynthetic assembly prior to installation at the cell surface. Two extracellular 6- O -endosulfatase enzymes (Sulf-1 and Sulf-2) can uniquely further edit mature HS and alter its interactions with GFs by removing specific sulfation motifs from their recognition sequence on HS. Despite being implicated as signaling regulators during development and in disease, the Sulfs have resisted structural characterization, and their substrate specificity and effects on GF interactions with HS are still poorly defined. Using a panel of PG-mimetics comprising compositionally-defined bioengineered recombinant HS (rHS) substrates in combination with GF binding and enzyme activity assays, we have discovered that Sulfs control GF-HS interactions through a combination of catalytic processing and competitive blocking of high affinity GF-binding sites, providing a new conceptual framework for understanding the functional impact of these enzymes in biological context. Although the contributions from each mechanism are both Sulf- and GF-dependent, the PG-mimetic platform allows for rapid analysis of these complex relationships. Significance Statement Cells rely on extracellular signals such as growth factors (GFs) to mediate critical biological functions. Membrane-associated proteins bearing negatively charged heparan sulfate (HS) sugar chains engage with GFs and present them to their receptors, which regulates their activity. Two extracellular sulfatase (Sulf) enzymes can edit HS and alter GF interactions and activity, although the precise mechanisms remain unclear. By using chemically defined HS-mimetics as probes, we have discovered that Sulfs can modulate HS by means of catalytic alterations and competitive blocking of GF-binding sites. These unique dual activities distinguish Sulfs from other enzymes and provide clues to their roles in development and disease.
Collapse
|
3
|
Maciej-Hulme ML, Leprince ACN, Lavin A, Guimond SE, Turnbull JE, Pelletier J, Yates EA, Powell AK, Skidmore MA. High sensitivity (zeptomole) detection of BODIPY-labelled heparan sulfate (HS) disaccharides by ion-paired RP-HPLC and LIF detection enables analysis of HS from mosquito midguts. ANALYTICAL METHODS : ADVANCING METHODS AND APPLICATIONS 2023; 15:1461-1469. [PMID: 36876452 PMCID: PMC10019443 DOI: 10.1039/d2ay01803a] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Accepted: 02/01/2023] [Indexed: 06/18/2023]
Abstract
The fine structure of heparan sulfate (HS), the glycosaminoglycan polysaccharide component of cell surface and extracellular matrix HS proteoglycans, coordinates the complex cell signalling processes that control homeostasis and drive development in multicellular animals. In addition, HS is involved in the infection of mammals by viruses, bacteria and parasites. The current detection limit for fluorescently labelled HS disaccharides (low femtomole; 10-15 mol), has effectively hampered investigations of HS composition in small, functionally-relevant populations of cells and tissues that may illuminate the structural requirements for infection and other biochemical processes. Here, an ultra-high sensitivity method is described that utilises a combination of reverse-phase HPLC, with tetraoctylammonium bromide (TOAB) as the ion-pairing reagent and laser-induced fluorescence detection of BODIPY-FL-labelled disaccharides. The method provides an unparalleled increase in the sensitivity of detection by ∼six orders of magnitude, enabling detection in the zeptomolar range (∼10-21 moles; <1000 labelled molecules). This facilitates determination of HS disaccharide compositional analysis from minute samples of selected tissues, as demonstrated by analysis of HS isolated from the midguts of Anopheles gambiae mosquitoes that was achieved without approaching the limit of detection.
Collapse
Affiliation(s)
- Marissa L Maciej-Hulme
- Centre for Glycoscience Research and Training, School of Life Sciences, Keele University, Staffordshire, ST5 5BG, UK.
| | - Anaëlle C N Leprince
- Centre for Glycoscience Research and Training, School of Life Sciences, Keele University, Staffordshire, ST5 5BG, UK.
- Université de Rennes 1, Rue du Thabor, 35065 Rennes Cedex, France
| | - Andre Lavin
- School of Pharmacy and Biomolecular Sciences, Liverpool John Moores University, Liverpool L3 3AF, UK
| | - Scott E Guimond
- Centre for Glycoscience Research and Training, School of Life Sciences, Keele University, Staffordshire, ST5 5BG, UK.
| | - Jeremy E Turnbull
- Centre for Glycoscience Research and Training, School of Life Sciences, Keele University, Staffordshire, ST5 5BG, UK.
| | - Julien Pelletier
- Centre for Glycoscience Research and Training, School of Life Sciences, Keele University, Staffordshire, ST5 5BG, UK.
| | - Edwin A Yates
- Centre for Glycoscience Research and Training, School of Life Sciences, Keele University, Staffordshire, ST5 5BG, UK.
- Department of Biochemistry and Systems Biology, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool L69 7ZB, UK
| | - Andrew K Powell
- School of Pharmacy and Biomolecular Sciences, Liverpool John Moores University, Liverpool L3 3AF, UK
| | - Mark A Skidmore
- Centre for Glycoscience Research and Training, School of Life Sciences, Keele University, Staffordshire, ST5 5BG, UK.
- Department of Biochemistry and Systems Biology, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool L69 7ZB, UK
| |
Collapse
|
4
|
Chemistry and Function of Glycosaminoglycans in the Nervous System. ADVANCES IN NEUROBIOLOGY 2023; 29:117-162. [DOI: 10.1007/978-3-031-12390-0_5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
|
5
|
Das S, Taylor K, Kozubek J, Sardell J, Gardner S. Genetic risk factors for ME/CFS identified using combinatorial analysis. J Transl Med 2022; 20:598. [PMID: 36517845 PMCID: PMC9749644 DOI: 10.1186/s12967-022-03815-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2022] [Accepted: 12/07/2022] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Myalgic encephalomyelitis/chronic fatigue syndrome (ME/CFS) is a debilitating chronic disease that lacks known pathogenesis, distinctive diagnostic criteria, and effective treatment options. Understanding the genetic (and other) risk factors associated with the disease would begin to help to alleviate some of these issues for patients. METHODS We applied both GWAS and the PrecisionLife combinatorial analytics platform to analyze ME/CFS cohorts from UK Biobank, including the Pain Questionnaire cohort, in a case-control design with 1000 cycles of fully random permutation. Results from this study were supported by a series of replication and cohort comparison experiments, including use of disjoint Verbal Interview CFS, post-viral fatigue syndrome and fibromyalgia cohorts also derived from UK Biobank, and compared results for overlap and reproducibility. RESULTS Combinatorial analysis revealed 199 SNPs mapping to 14 genes that were significantly associated with 91% of the cases in the ME/CFS population. These SNPs were found to stratify by shared cases into 15 clusters (communities) made up of 84 high-order combinations of between 3 and 5 SNPs. p-values for these communities range from 2.3 × 10-10 to 1.6 × 10-72. Many of the genes identified are linked to the key cellular mechanisms hypothesized to underpin ME/CFS, including vulnerabilities to stress and/or infection, mitochondrial dysfunction, sleep disturbance and autoimmune development. We identified 3 of the critical SNPs replicated in the post-viral fatigue syndrome cohort and 2 SNPs replicated in the fibromyalgia cohort. We also noted similarities with genes associated with multiple sclerosis and long COVID, which share some symptoms and potentially a viral infection trigger with ME/CFS. CONCLUSIONS This study provides the first detailed genetic insights into the pathophysiological mechanisms underpinning ME/CFS and offers new approaches for better diagnosis and treatment of patients.
Collapse
Affiliation(s)
- Sayoni Das
- PrecisionLife Ltd, Long Hanborough, Oxford, UK
| | | | | | | | | |
Collapse
|
6
|
Caldwell ALM, Sancho L, Deng J, Bosworth A, Miglietta A, Diedrich JK, Shokhirev MN, Allen NJ. Aberrant astrocyte protein secretion contributes to altered neuronal development in multiple models of neurodevelopmental disorders. Nat Neurosci 2022; 25:1163-1178. [PMID: 36042312 PMCID: PMC10395413 DOI: 10.1038/s41593-022-01150-1] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Accepted: 07/20/2022] [Indexed: 01/01/2023]
Abstract
Astrocytes negatively impact neuronal development in many models of neurodevelopmental disorders (NDs); however, how they do this, and if mechanisms are shared across disorders, is not known. In this study, we developed a cell culture system to ask how astrocyte protein secretion and gene expression change in three mouse models of genetic NDs (Rett, Fragile X and Down syndromes). ND astrocytes increase release of Igfbp2, a secreted inhibitor of insulin-like growth factor (IGF). IGF rescues neuronal deficits in many NDs, and we found that blocking Igfbp2 partially rescues inhibitory effects of Rett syndrome astrocytes, suggesting that increased astrocyte Igfbp2 contributes to decreased IGF signaling in NDs. We identified that increased BMP signaling is upstream of protein secretion changes, including Igfbp2, and blocking BMP signaling in Fragile X and Rett syndrome astrocytes reverses inhibitory effects on neurite outgrowth. This work provides a resource of astrocyte-secreted proteins in health and ND models and identifies novel targets for intervention in diverse NDs.
Collapse
Affiliation(s)
- Alison L M Caldwell
- Molecular Neurobiology Laboratory, Salk Institute for Biological Studies, La Jolla, CA, USA
- Neurosciences Graduate Program, University of California, San Diego, La Jolla, CA, USA
| | - Laura Sancho
- Molecular Neurobiology Laboratory, Salk Institute for Biological Studies, La Jolla, CA, USA
| | - James Deng
- Molecular Neurobiology Laboratory, Salk Institute for Biological Studies, La Jolla, CA, USA
- Neurosciences Graduate Program, University of California, San Diego, La Jolla, CA, USA
| | - Alexandra Bosworth
- Molecular Neurobiology Laboratory, Salk Institute for Biological Studies, La Jolla, CA, USA
- Neurosciences Graduate Program, University of California, San Diego, La Jolla, CA, USA
| | - Audrey Miglietta
- Molecular Neurobiology Laboratory, Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Jolene K Diedrich
- Mass Spectrometry Core, Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Maxim N Shokhirev
- Razavi Newman Integrative Genomics and Bioinformatics Core, Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Nicola J Allen
- Molecular Neurobiology Laboratory, Salk Institute for Biological Studies, La Jolla, CA, USA.
| |
Collapse
|
7
|
Fawcett JW, Kwok JCF. Proteoglycan Sulphation in the Function of the Mature Central Nervous System. Front Integr Neurosci 2022; 16:895493. [PMID: 35712345 PMCID: PMC9195417 DOI: 10.3389/fnint.2022.895493] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2022] [Accepted: 04/21/2022] [Indexed: 11/13/2022] Open
Abstract
Chondroitin sulphate and heparan sulphate proteoglycans (CSPGS and HSPGs) are found throughout the central nervous system (CNS). CSPGs are ubiquitous in the diffuse extracellular matrix (ECM) between cells and are a major component of perineuronal nets (PNNs), the condensed ECM present around some neurons. HSPGs are more associated with the surface of neurons and glia, with synapses and in the PNNs. Both CSPGs and HSPGs consist of a protein core to which are attached repeating disaccharide chains modified by sulphation at various positions. The sequence of sulphation gives the chains a unique structure and local charge density. These sulphation codes govern the binding properties and biological effects of the proteoglycans. CSPGs are sulphated along their length, the main forms being 6- and 4-sulphated. In general, the chondroitin 4-sulphates are inhibitory to cell attachment and migration, while chondroitin 6-sulphates are more permissive. HSPGs tend to be sulphated in isolated motifs with un-sulphated regions in between. The sulphation patterns of HS motifs and of CS glycan chains govern their binding to the PTPsigma receptor and binding of many effector molecules to the proteoglycans, such as growth factors, morphogens, and molecules involved in neurodegenerative disease. Sulphation patterns change as a result of injury, inflammation and ageing. For CSPGs, attention has focussed on PNNs and their role in the control of plasticity and memory, and on the soluble CSPGs upregulated in glial scar tissue that can inhibit axon regeneration. HSPGs have key roles in development, regulating cell migration and axon growth. In the adult CNS, they have been associated with tau aggregation and amyloid-beta processing, synaptogenesis, growth factor signalling and as a component of the stem cell niche. These functions of CSPGs and HSPGs are strongly influenced by the pattern of sulphation of the glycan chains, the sulphation code. This review focuses on these sulphation patterns and their effects on the function of the mature CNS.
Collapse
Affiliation(s)
- James W. Fawcett
- Department of Clinical Neurosciences, John van Geest Centre for Brain Repair, University of Cambridge, Cambridge, United Kingdom
- Centre for Reconstructive Neuroscience, Institute for Experimental Medicine Czech Academy of Science (CAS), Prague, Czechia
| | - Jessica C. F. Kwok
- Centre for Reconstructive Neuroscience, Institute for Experimental Medicine Czech Academy of Science (CAS), Prague, Czechia
- Faculty of Biological Sciences, School of Biomedical Sciences, University of Leeds, Leeds, United Kingdom
| |
Collapse
|
8
|
Mashima R, Okuyama T, Ohira M. Physiology and Pathophysiology of Heparan Sulfate in Animal Models: Its Biosynthesis and Degradation. Int J Mol Sci 2022; 23:1963. [PMID: 35216081 PMCID: PMC8876164 DOI: 10.3390/ijms23041963] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Revised: 02/05/2022] [Accepted: 02/08/2022] [Indexed: 12/17/2022] Open
Abstract
Heparan sulfate (HS) is a type of glycosaminoglycan that plays a key role in a variety of biological functions in neurology, skeletal development, immunology, and tumor metastasis. Biosynthesis of HS is initiated by a link of xylose to Ser residue of HS proteoglycans, followed by the formation of a linker tetrasaccharide. Then, an extension reaction of HS disaccharide occurs through polymerization of many repetitive units consisting of iduronic acid and N-acetylglucosamine. Subsequently, several modification reactions take place to complete the maturation of HS. The sulfation positions of N-, 2-O-, 6-O-, and 3-O- are all mediated by specific enzymes that may have multiple isozymes. C5-epimerization is facilitated by the epimerase enzyme that converts glucuronic acid to iduronic acid. Once these enzymatic reactions have been completed, the desulfation reaction further modifies HS. Apart from HS biosynthesis, the degradation of HS is largely mediated by the lysosome, an intracellular organelle with acidic pH. Mucopolysaccharidosis is a genetic disorder characterized by an accumulation of glycosaminoglycans in the body associated with neuronal, skeletal, and visceral disorders. Genetically modified animal models have significantly contributed to the understanding of the in vivo role of these enzymes. Their role and potential link to diseases are also discussed.
Collapse
Affiliation(s)
- Ryuichi Mashima
- Department of Clinical Laboratory Medicine, National Center for Child Health and Development, 2-10-1 Okura, Setagaya-ku, Tokyo 157-8535, Japan; (T.O.); (M.O.)
| | | | | |
Collapse
|
9
|
Association between maternal depression during pregnancy and newborn DNA methylation. Transl Psychiatry 2021; 11:572. [PMID: 34750344 PMCID: PMC8576002 DOI: 10.1038/s41398-021-01697-w] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Revised: 09/22/2021] [Accepted: 10/22/2021] [Indexed: 01/28/2023] Open
Abstract
Around 15-65% of women globally experience depression during pregnancy, prevalence being particularly high in low- and middle-income countries. Prenatal depression has been associated with adverse birth and child development outcomes. DNA methylation (DNAm) may aid in understanding this association. In this project, we analyzed associations between prenatal depression and DNAm from cord blood from participants of the South African Drakenstein Child Health Study. We examined DNAm in an epigenome-wide association study (EWAS) of 248 mother-child pairs. DNAm was measured using the Infinium MethylationEPIC (N = 145) and the Infinium HumanMethylation450 (N = 103) arrays. Prenatal depression scores, obtained with the Edinburgh Postnatal Depression Scale (EPDS) and the Beck Depression Inventory-II (BDI-II), were analyzed as continuous and dichotomized variables. We used linear robust models to estimate associations between depression and newborn DNAm, adjusted for measured (smoking status, household income, sex, preterm birth, cell type proportions, and genetic principal components) and unmeasured confounding using Cate and Bacon algorithms. Bonferroni correction was used to adjust for multiple testing. DMRcate and dmrff were used to test for differentially methylated regions (DMRs). Differential DNAm was significantly associated with BDI-II variables, in cg16473797 (Δ beta = -1.10E-02, p = 6.87E-08), cg23262030 (Δ beta per BDI-II total IQR = 1.47E-03, p = 1.18E-07), and cg04859497 (Δ beta = -6.42E-02, p = 1.06E-09). Five DMRs were associated with at least two depression variables. Further studies are needed to replicate these findings and investigate their biological impact.
Collapse
|
10
|
Nanus DE, Badoume A, Wijesinghe SN, Halsey AM, Hurley P, Ahmed Z, Botchu R, Davis ET, Lindsay MA, Jones SW. Synovial tissue from sites of joint pain in knee osteoarthritis patients exhibits a differential phenotype with distinct fibroblast subsets. EBioMedicine 2021; 72:103618. [PMID: 34628351 PMCID: PMC8511845 DOI: 10.1016/j.ebiom.2021.103618] [Citation(s) in RCA: 59] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Revised: 09/01/2021] [Accepted: 09/22/2021] [Indexed: 02/02/2023] Open
Abstract
BACKGROUND Synovial inflammation is associated with pain severity in patients with knee osteoarthritis (OA). The aim here was to determine in a population with knee OA, whether synovial tissue from areas associated with pain exhibited different synovial fibroblast subsets, compared to synovial tissue from sites not associated with pain. A further aim was to compare differences between early and end-stage disease synovial fibroblast subsets. METHODS Patients with early knee OA (n = 29) and end-stage knee OA (n = 22) were recruited. Patient reported pain was recorded by questionnaire and using an anatomical knee pain map. Proton density fat suppressed MRI axial and sagittal sequences were analysed and scored for synovitis. Synovial tissue was obtained from the medial and lateral parapatellar and suprapatellar sites. Fibroblast single cell RNA sequencing was performed using Chromium 10X and analysed using Seurat. Transcriptomes were functionally characterised using Ingenuity Pathway Analysis and the effect of fibroblast secretome on neuronal growth assessed using rat DRGN. FINDINGS Parapatellar synovitis was significantly associated with the pattern of patient-reported pain in knee OA patients. Synovial tissue from sites of patient-reported pain exhibited a differential transcriptomic phenotype, with distinct synovial fibroblast subsets in early OA and end-stage OA. Functional pathway analysis revealed that synovial tissue and fibroblast subsets from painful sites promoted fibrosis, inflammation and the growth and activity of neurons. The secretome of fibroblasts from early OA painful sites induced greater survival and neurite outgrowth in dissociated adult rodent dorsal root ganglion neurons. INTERPRETATION Sites of patient-reported pain in knee OA exhibit a different synovial tissue phenotype and distinct synovial fibroblast subsets. Further interrogation of these fibroblast pathotypes will increase our understanding of the role of synovitis in OA joint pain and provide a rationale for the therapeutic targeting of fibroblast subsets to alleviate pain in patients. FUNDING This study was funded by Versus Arthritis, UK (21530; 21812).
Collapse
Affiliation(s)
- Dominika E Nanus
- Institute of Inflammation and Ageing, MRC-ARUK Centre for Musculoskeletal Ageing Research, University of Birmingham, Birmingham B15 2TT, United Kingdom
| | - Amel Badoume
- Department of Pharmacy and Pharmacology, University of Bath, Claverton Down, Bath BA2 7AY, United Kingdom
| | - Susanne N Wijesinghe
- Institute of Inflammation and Ageing, MRC-ARUK Centre for Musculoskeletal Ageing Research, University of Birmingham, Birmingham B15 2TT, United Kingdom
| | - Andrea M Halsey
- Neuroscience and Ophthalmology, Institute of Inflammation and Ageing, University of Birmingham, Birmingham B15 2TT, United Kingdom
| | - Patrick Hurley
- Institute of Inflammation and Ageing, MRC-ARUK Centre for Musculoskeletal Ageing Research, University of Birmingham, Birmingham B15 2TT, United Kingdom
| | - Zubair Ahmed
- Neuroscience and Ophthalmology, Institute of Inflammation and Ageing, University of Birmingham, Birmingham B15 2TT, United Kingdom,Centre for Trauma Sciences Research, University of Birmingham, Birmingham B15 2TT, United Kingdom
| | - Rajesh Botchu
- The Royal Orthopaedic Hospital, Birmingham B31 2AP, United Kingdom
| | - Edward T Davis
- Institute of Inflammation and Ageing, MRC-ARUK Centre for Musculoskeletal Ageing Research, University of Birmingham, Birmingham B15 2TT, United Kingdom,The Royal Orthopaedic Hospital, Birmingham B31 2AP, United Kingdom
| | - Mark A Lindsay
- Institute of Inflammation and Ageing, MRC-ARUK Centre for Musculoskeletal Ageing Research, University of Birmingham, Birmingham B15 2TT, United Kingdom
| | - Simon W Jones
- Institute of Inflammation and Ageing, MRC-ARUK Centre for Musculoskeletal Ageing Research, University of Birmingham, Birmingham B15 2TT, United Kingdom,Corresponding author.
| |
Collapse
|
11
|
Miya K, Keino-Masu K, Okada T, Kobayashi K, Masu M. Expression of Heparan Sulfate Endosulfatases in the Adult Mouse Brain: Co-expression of Sulf1 and Dopamine D1/D2 Receptors. Front Neuroanat 2021; 15:726718. [PMID: 34489650 PMCID: PMC8417564 DOI: 10.3389/fnana.2021.726718] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2021] [Accepted: 07/26/2021] [Indexed: 11/13/2022] Open
Abstract
The heparan sulfate 6-O-endosulfatases, Sulfatase 1 (Sulf1), and Sulfatase 2 (Sulf2), are extracellular enzymes that regulate cellular signaling by removing 6-O-sulfate from the heparan sulfate chain. Although previous studies have revealed that Sulfs are essential for normal development, their functions in the adult brain remain largely unknown. To gain insight into their neural functions, we used in situ hybridization to systematically examine Sulf1/2 mRNA expression in the adult mouse brain. Sulf1 and Sulf2 mRNAs showed distinct expression patterns, which is in contrast to their overlapping expression in the embryonic brain. In addition, we found that Sulf1 was distinctly expressed in the nucleus accumbens shell, the posterior tail of the striatum, layer 6 of the cerebral cortex, and the paraventricular nucleus of the thalamus, all of which are target areas of dopaminergic projections. Using double-labeling techniques, we showed that Sulf1-expressing cells in the above regions coincided with cells expressing the dopamine D1 and/or D2 receptor. These findings implicate possible roles of Sulf1 in modulation of dopaminergic transmission and dopamine-mediated behaviors.
Collapse
Affiliation(s)
- Ken Miya
- Graduate School of Comprehensive Human Sciences, University of Tsukuba, Tsukuba, Japan.,Department of Molecular Neurobiology, Division of Biomedical Science, Faculty of Medicine, University of Tsukuba, Tsukuba, Japan
| | - Kazuko Keino-Masu
- Graduate School of Comprehensive Human Sciences, University of Tsukuba, Tsukuba, Japan.,Department of Molecular Neurobiology, Division of Biomedical Science, Faculty of Medicine, University of Tsukuba, Tsukuba, Japan
| | - Takuya Okada
- Graduate School of Comprehensive Human Sciences, University of Tsukuba, Tsukuba, Japan.,Department of Molecular Neurobiology, Division of Biomedical Science, Faculty of Medicine, University of Tsukuba, Tsukuba, Japan
| | - Kenta Kobayashi
- Section of Viral Vector Development, Center for Genetic Analysis of Behavior, National Institute for Physiological Sciences, National Institutes of Natural Sciences, Okazaki, Japan
| | - Masayuki Masu
- Graduate School of Comprehensive Human Sciences, University of Tsukuba, Tsukuba, Japan.,Department of Molecular Neurobiology, Division of Biomedical Science, Faculty of Medicine, University of Tsukuba, Tsukuba, Japan
| |
Collapse
|
12
|
Maiya R, Pomrenze MB, Tran T, Tiwari GR, Beckham A, Paul MT, Mayfield RD, Messing RO. Differential regulation of alcohol consumption and reward by the transcriptional cofactor LMO4. Mol Psychiatry 2021; 26:2175-2186. [PMID: 32144357 PMCID: PMC7558853 DOI: 10.1038/s41380-020-0706-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/14/2019] [Revised: 02/20/2020] [Accepted: 02/25/2020] [Indexed: 01/04/2023]
Abstract
Repeated alcohol exposure leads to changes in gene expression that are thought to underlie the transition from moderate to excessive drinking. However, the mechanisms by which these changes are integrated into a maladaptive response that leads to alcohol dependence are not well understood. One mechanism could involve the recruitment of transcriptional co-regulators that bind and modulate the activity of transcription factors. Our results indicate that the transcriptional regulator LMO4 is one such candidate regulator. Lmo4-deficient mice (Lmo4gt/+) consumed significantly more and showed enhanced preference for alcohol in a 24 h intermittent access drinking procedure. shRNA-mediated knockdown of Lmo4 in the nucleus accumbens enhanced alcohol consumption, whereas knockdown in the basolateral amygdala (BLA) decreased alcohol consumption and reduced conditioned place preference for alcohol. To ascertain the molecular mechanisms that underlie these contrasting phenotypes, we carried out unbiased transcriptome profiling of these two brain regions in wild type and Lmo4gt/+ mice. Our results revealed that the transcriptional targets of LMO4 are vastly different between the two brain regions, which may explain the divergent phenotypes observed upon Lmo4 knockdown. Bioinformatic analyses revealed that Oprk1 and genes related to the extracellular matrix (ECM) are important transcriptional targets of LMO4 in the BLA. Chromatin immunoprecipitation revealed that LMO4 bound Oprk1 promoter elements. Consistent with these results, disruption of the ECM or infusion of norbinaltorphimine, a selective kappa opioid receptor antagonist, in the BLA reduced alcohol consumption. Hence our results indicate that an LMO4-regulated transcriptional network regulates alcohol consumption in the BLA.
Collapse
Affiliation(s)
- Rajani Maiya
- Department of Neuroscience, The University of Texas at Austin, Austin, TX, 78712, USA. .,Waggoner Center for Alcohol and Addiction Research, The University of Texas at Austin, Austin, TX, 78712, USA. .,Department of Neurology, The University of Texas at Austin, Austin, TX, 78712, USA.
| | - Matthew B. Pomrenze
- Department of Neuroscience, The University of Texas at Austin, Austin, TX 78712, USA,Waggoner Center for Alcohol and Addiction Research, The University of Texas at Austin, Austin, TX 78712, USA
| | - Thi Tran
- Department of Neuroscience, The University of Texas at Austin, Austin, TX 78712, USA
| | - Gayatri R. Tiwari
- Waggoner Center for Alcohol and Addiction Research, The University of Texas at Austin, Austin, TX 78712, USA
| | - Andrea Beckham
- Department of Neuroscience, The University of Texas at Austin, Austin, TX 78712, USA
| | - Madison T. Paul
- Department of Neuroscience, The University of Texas at Austin, Austin, TX 78712, USA
| | - R. Dayne Mayfield
- Department of Neuroscience, The University of Texas at Austin, Austin, TX 78712, USA,Waggoner Center for Alcohol and Addiction Research, The University of Texas at Austin, Austin, TX 78712, USA
| | - Robert O. Messing
- Department of Neuroscience, The University of Texas at Austin, Austin, TX 78712, USA,Waggoner Center for Alcohol and Addiction Research, The University of Texas at Austin, Austin, TX 78712, USA,Department of Neurology, The University of Texas at Austin, Austin, TX 78712, USA
| |
Collapse
|
13
|
Zeng F, Liu Y, Ouyang Q, Sun Z, Zhang K, Li X, Liu Y. Rs3802278 in 3'-UTR of SULF1 associated with platinum resistance and survival in Chinese epithelial ovarian cancer patients. J Chemother 2021; 33:564-569. [PMID: 34029511 DOI: 10.1080/1120009x.2021.1913702] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
Ovarian cancer is the leading cause of death from gynecologic cancers, but platinum resistance remains a major obstacle in the chemotherapy of ovarian cancer. This study aims to examine the role of polymorphisms in sulfatase 1 (SULF1) in platinum resistance and survival in advanced epithelial ovarian cancer (EOC) patients. We genotyped 12 SNPs of SULF1 in 195 EOC patients treated with platinum using MassARRAY method and evaluated the association between the SNPs and platinum response. SULF1 rs3802278 was marginal significantly associated with platinum resistance in recessive model with p value of 0.055. The patients with SULF1 rs3802278 AA were more resistant to platinum-based chemotherapy comparing to those with AG/GG genotype (OR: 2.317, 95%CI: 0.982 ∼ 5.465). In survival analysis, rs3802278 was significantly associated with both of PFS and OS after adjusted by FIGO stage and age. Patients with AA genotypes showed a shorter PFS and OS than with AG/GG genotypes (median PFS: 15 months vs. 21 months, p = 0.010, HR = 1.876, 95%CI: 1.165-3.022; median OS: 42 months vs. 73 months, p = 0.031, HR = 1.928, 95%CI: 1.061-3.504). SULF1 rs3802278 may serve as a potential candidate biomarker for the prediction of platinum resistance and prognosis in Chinese EOC patients.
Collapse
Affiliation(s)
- Feiyue Zeng
- Department of Radiology, Xiangya Hospital, Central South University, Changsha, Hunan, P. R. China
| | - Yujie Liu
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, Hunan, P. R. China.,Institute of Clinical Pharmacology, Central South University, Hunan Key Laboratory of Pharmacogenetics, Changsha, Hunan, P. R. China.,Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, Changsha, Hunan, P. R. China.,National Clinical Research Center for Geriatric Disorders, Changsha, Hunan, P. R. China
| | - Qianying Ouyang
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, Hunan, P. R. China.,Institute of Clinical Pharmacology, Central South University, Hunan Key Laboratory of Pharmacogenetics, Changsha, Hunan, P. R. China.,Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, Changsha, Hunan, P. R. China.,National Clinical Research Center for Geriatric Disorders, Changsha, Hunan, P. R. China
| | - Zeen Sun
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, Hunan, P. R. China.,Institute of Clinical Pharmacology, Central South University, Hunan Key Laboratory of Pharmacogenetics, Changsha, Hunan, P. R. China.,Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, Changsha, Hunan, P. R. China.,National Clinical Research Center for Geriatric Disorders, Changsha, Hunan, P. R. China
| | - Keqiang Zhang
- Hunan Provincial Tumor Hospital, The Affiliated Tumor Hospital of Xiangya Medical School of Central South University, Changsha, Hunan, P. R. China
| | - Xi Li
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, Hunan, P. R. China.,Institute of Clinical Pharmacology, Central South University, Hunan Key Laboratory of Pharmacogenetics, Changsha, Hunan, P. R. China.,Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, Changsha, Hunan, P. R. China.,National Clinical Research Center for Geriatric Disorders, Changsha, Hunan, P. R. China
| | - Yingzi Liu
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, Hunan, P. R. China.,Institute of Clinical Pharmacology, Central South University, Hunan Key Laboratory of Pharmacogenetics, Changsha, Hunan, P. R. China.,Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, Changsha, Hunan, P. R. China.,National Clinical Research Center for Geriatric Disorders, Changsha, Hunan, P. R. China
| |
Collapse
|
14
|
Silva TP, Sousa-Luís R, Fernandes TG, Bekman EP, Rodrigues CAV, Vaz SH, Moreira LM, Hashimura Y, Jung S, Lee B, Carmo-Fonseca M, Cabral JMS. Transcriptome profiling of human pluripotent stem cell-derived cerebellar organoids reveals faster commitment under dynamic conditions. Biotechnol Bioeng 2021; 118:2781-2803. [PMID: 33871054 DOI: 10.1002/bit.27797] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Revised: 03/30/2021] [Accepted: 04/14/2021] [Indexed: 12/14/2022]
Abstract
Human-induced pluripotent stem cells (iPSCs) have great potential for disease modeling. However, generating iPSC-derived models to study brain diseases remains a challenge. In particular, the ability to recapitulate cerebellar development in vitro is still limited. We presented a reproducible and scalable production of cerebellar organoids by using the novel single-use Vertical-Wheel bioreactors, in which functional cerebellar neurons were obtained. Here, we evaluate the global gene expression profiles by RNA sequencing (RNA-seq) across cerebellar differentiation, demonstrating a faster cerebellar commitment in this novel dynamic differentiation protocol. Furthermore, transcriptomic profiles suggest a significant enrichment of extracellular matrix (ECM) in dynamic-derived cerebellar organoids, which can better mimic the neural microenvironment and support a consistent neuronal network. Thus, an efficient generation of organoids with cerebellar identity was achieved for the first time in a continuous process using a dynamic system without the need of organoids encapsulation in ECM-based hydrogels, allowing the possibility of large-scale production and application in high-throughput processes. The presence of factors that favors angiogenesis onset was also detected in dynamic conditions, which can enhance functional maturation of cerebellar organoids. We anticipate that large-scale production of cerebellar organoids may help developing models for drug screening, toxicological tests, and studying pathological pathways involved in cerebellar degeneration.
Collapse
Affiliation(s)
- Teresa P Silva
- Department of Bioengineering and Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Lisboa, Portugal.,Associate Laboratory i4HB - Institute for Health and Bioeconomy, Instituto Superior Técnico, Universidade de Lisboa, Lisboa, Portugal.,Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
| | - Rui Sousa-Luís
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
| | - Tiago G Fernandes
- Department of Bioengineering and Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Lisboa, Portugal.,Associate Laboratory i4HB - Institute for Health and Bioeconomy, Instituto Superior Técnico, Universidade de Lisboa, Lisboa, Portugal
| | - Evguenia P Bekman
- Department of Bioengineering and Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Lisboa, Portugal.,Associate Laboratory i4HB - Institute for Health and Bioeconomy, Instituto Superior Técnico, Universidade de Lisboa, Lisboa, Portugal.,Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
| | - Carlos A V Rodrigues
- Department of Bioengineering and Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Lisboa, Portugal.,Associate Laboratory i4HB - Institute for Health and Bioeconomy, Instituto Superior Técnico, Universidade de Lisboa, Lisboa, Portugal
| | - Sandra H Vaz
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal.,Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal, Portugal
| | - Leonilde M Moreira
- Department of Bioengineering and Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Lisboa, Portugal.,Associate Laboratory i4HB - Institute for Health and Bioeconomy, Instituto Superior Técnico, Universidade de Lisboa, Lisboa, Portugal
| | | | | | - Brian Lee
- PBS Biotech, Camarillo, California, USA
| | - Maria Carmo-Fonseca
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
| | - Joaquim M S Cabral
- Department of Bioengineering and Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Lisboa, Portugal.,Associate Laboratory i4HB - Institute for Health and Bioeconomy, Instituto Superior Técnico, Universidade de Lisboa, Lisboa, Portugal
| |
Collapse
|
15
|
Pérez-López N, Martín C, García B, Solís-Hernández MP, Rodríguez D, Alcalde I, Merayo J, Fernández-Vega I, Quirós LM. Alterations in the Expression of the Genes Responsible for the Synthesis of Heparan Sulfate in Brains With Alzheimer Disease. J Neuropathol Exp Neurol 2021; 80:446-456. [PMID: 33779723 DOI: 10.1093/jnen/nlab028] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
The saccharide chains of heparan sulfate appear to be involved in several aspects Alzheimer disease (AD) pathogenesis. Their structural complexity is due to the expression of different isoenzymes. We studied the differential transcription of heparan sulfate chain biosynthesis in AD brains, analyzing different brain regions in patients with different extents of AD pathology. The transcriptomic study was performed by RT-PCR using samples of amygdala, anterior hippocampus, posterior hippocampus, claustrum, calcarine fissure, globus pallidus and cerebellum from patients with mild, moderate, or severe AD, as well as healthy individuals. Certain heparan sulfate epitopes were also detected by immunohistochemistry. Several genes, across all stages of heparan sulfate synthesis, showed altered transcription in different brain regions of AD patients. The numbers of alterations were greater in in moderate versus mild AD patients. In severe patients, there were fewer alterations in genes related to early stages of biosynthesis, and overexpression of genes involved in late stages. The alterations correlated with progressive brain atrophy, although alterations were more common in the cerebellum. Detection of some heparan sulfate epitopes by immunohistochemistry was consistent with previous studies. In conclusion, transcriptional alterations in the biosynthetic genes of heparan sulfate depend on the brain region and the degree of AD pathology.
Collapse
Affiliation(s)
- Natalia Pérez-López
- From the Instituto Universitario Fernández-Vega, University of Oviedo, Oviedo, Spain.,Instituto de Investigación Sanitaria del Principado de Asturias, Oviedo, Spain
| | - Carla Martín
- From the Instituto Universitario Fernández-Vega, University of Oviedo, Oviedo, Spain.,Instituto de Investigación Sanitaria del Principado de Asturias, Oviedo, Spain.,Department of Functional Biology, University of Oviedo, Oviedo, Spain
| | - Beatriz García
- Department of Functional Biology, University of Oviedo, Oviedo, Spain
| | | | - David Rodríguez
- Department of Biochemistry and Molecular Biology, University of Oviedo, Oviedo, Spain.,Instituto Universitario de Oncología del Principado de Asturias, University of Oviedo, Oviedo, Spain
| | - Ignacio Alcalde
- From the Instituto Universitario Fernández-Vega, University of Oviedo, Oviedo, Spain.,Instituto de Investigación Sanitaria del Principado de Asturias, Oviedo, Spain
| | - Jesús Merayo
- From the Instituto Universitario Fernández-Vega, University of Oviedo, Oviedo, Spain.,Instituto de Investigación Sanitaria del Principado de Asturias, Oviedo, Spain.,Department of Surgery and Medical-surgical Specialties, University of Oviedo, Oviedo, Spain
| | - Iván Fernández-Vega
- From the Instituto Universitario Fernández-Vega, University of Oviedo, Oviedo, Spain.,Instituto de Investigación Sanitaria del Principado de Asturias, Oviedo, Spain.,Department of Surgery and Medical-surgical Specialties, University of Oviedo, Oviedo, Spain.,Department of Pathology, Hospital Universitario Central de Asturias, Oviedo, Spain
| | - Luis M Quirós
- From the Instituto Universitario Fernández-Vega, University of Oviedo, Oviedo, Spain.,Instituto de Investigación Sanitaria del Principado de Asturias, Oviedo, Spain.,Department of Functional Biology, University of Oviedo, Oviedo, Spain
| |
Collapse
|
16
|
Sytnyk V, Leshchyns'ka I, Schachner M. Neural glycomics: the sweet side of nervous system functions. Cell Mol Life Sci 2021; 78:93-116. [PMID: 32613283 PMCID: PMC11071817 DOI: 10.1007/s00018-020-03578-9] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Revised: 06/06/2020] [Accepted: 06/22/2020] [Indexed: 02/07/2023]
Abstract
The success of investigations on the structure and function of the genome (genomics) has been paralleled by an equally awesome progress in the analysis of protein structure and function (proteomics). We propose that the investigation of carbohydrate structures that go beyond a cell's metabolism is a rapidly developing frontier in our expanding knowledge on the structure and function of carbohydrates (glycomics). No other functional system appears to be suited as well as the nervous system to study the functions of glycans, which had been originally characterized outside the nervous system. In this review, we describe the multiple studies on the functions of LewisX, the human natural killer cell antigen-1 (HNK-1), as well as oligomannosidic and sialic (neuraminic) acids. We attempt to show the sophistication of these structures in ontogenetic development, synaptic function and plasticity, and recovery from trauma, with a view on neurodegeneration and possibilities to ameliorate deterioration. In view of clinical applications, we emphasize the need for glycomimetic small organic compounds which surpass the usefulness of natural glycans in that they are metabolically more stable, more parsimonious to synthesize or isolate, and more advantageous for therapy, since many of them pass the blood brain barrier and are drug-approved for treatments other than those in the nervous system, thus allowing a more ready access for application in neurological diseases. We describe the isolation of such mimetic compounds using not only Western NIH, but also traditional Chinese medical libraries. With this review, we hope to deepen the interests in this exciting field.
Collapse
Affiliation(s)
- Vladimir Sytnyk
- School of Biotechnology and Biomolecular Sciences, The University of New South Wales, Sydney, NSW, Australia.
| | - Iryna Leshchyns'ka
- School of Biotechnology and Biomolecular Sciences, The University of New South Wales, Sydney, NSW, Australia
| | - Melitta Schachner
- Center for Neuroscience, Shantou University Medical College, 22 Xin Ling Road, Shantou, 515041, Guangdong, China
- Department of Cell Biology and Neuroscience, Keck Center for Collaborative Neuroscience, Rutgers University, 604 Allison Road, Piscataway, NJ, 08854, USA
| |
Collapse
|
17
|
Heparan Sulfate Proteoglycans Biosynthesis and Post Synthesis Mechanisms Combine Few Enzymes and Few Core Proteins to Generate Extensive Structural and Functional Diversity. Molecules 2020; 25:molecules25184215. [PMID: 32937952 PMCID: PMC7570499 DOI: 10.3390/molecules25184215] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Revised: 09/08/2020] [Accepted: 09/09/2020] [Indexed: 02/06/2023] Open
Abstract
Glycosylation is a common and widespread post-translational modification that affects a large majority of proteins. Of these, a small minority, about 20, are specifically modified by the addition of heparan sulfate, a linear polysaccharide from the glycosaminoglycan family. The resulting molecules, heparan sulfate proteoglycans, nevertheless play a fundamental role in most biological functions by interacting with a myriad of proteins. This large functional repertoire stems from the ubiquitous presence of these molecules within the tissue and a tremendous structural variety of the heparan sulfate chains, generated through both biosynthesis and post synthesis mechanisms. The present review focusses on how proteoglycans are “gagosylated” and acquire structural complexity through the concerted action of Golgi-localized biosynthesis enzymes and extracellular modifying enzymes. It examines, in particular, the possibility that these enzymes form complexes of different modes of organization, leading to the synthesis of various oligosaccharide sequences.
Collapse
|
18
|
Xun C, Ge L, Tang F, Wang L, Zhuo Y, Long L, Qi J, Hu L, Duan D, Chen P, Lu M. Insight into the proteomic profiling of exosomes secreted by human OM-MSCs reveals a new potential therapy. Biomed Pharmacother 2020; 131:110584. [PMID: 32841894 DOI: 10.1016/j.biopha.2020.110584] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Revised: 07/05/2020] [Accepted: 07/25/2020] [Indexed: 01/08/2023] Open
Abstract
Mesenchymal stromal cells (MSCs) have been used for the treatment of neuronal injury and neurodegenerative diseases. Their underlying mechanism may involve increased secretion of paracrine factors, which promotes tissue repair. Presently, exosomes have been regarded as important components of paracrine secretion and paracrine factors. MSC exosomes represent a promising opportunity to develop novel cell-free therapy approaches. In this study, exosomes from nasal olfactory mucosa MSCs (OM-MSCs) were extracted and purified using ultracentrifugation, resulting in exosome diameters of 40-130 nm. Similar to other exosomes, OM-MSC exosomes were CD63- and CD81-positive and calnexin-negative. Functionally, OM-MSC exosomes promoted human brain microvascular endothelial cell (HBMEC) proliferation and migration. The present study analyzed the OM-MSC exosome paracrine proteome. A total of 304 exosome-associated proteins were identified by LC-MS/MS, including plasminogen activator inhibitor 1 (SERPINE 1), insulin-like growth factor binding protein family members (IGFBP 4 and 5), epidermal growth factor receptor (EGFR), neurogenic locus notch homolog protein 2 (NOTCH 2), apolipoprotein E (APOE), and heat shock protein HSP90-beta (HSP90AB1). These molecules are known to be important in neurotrophic, angiogenesis, cell growth, differentiation, apoptosis, and inflammation and are highly correlated with the mechanism of tissue repair and neural restoration. These observations may provide a basis for further evaluation of OM-MSC exosome potential as a novel therapeutic modality.
Collapse
Affiliation(s)
- Chengfeng Xun
- The National & Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Sciences, Hunan Normal University, Changsha Hunan 410081, China
| | - Lite Ge
- The National & Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Sciences, Hunan Normal University, Changsha Hunan 410081, China; Department of Neurology, Second Xiangya Hospital, Central South University, Changsha Hunan, 410011, China; Hunan Provincical Key Laboratory of Neurorestoratology, the Second Affiliated Hospital of Hunan Normal University, Changsha Hunan, 410003, China
| | - Feng Tang
- The National & Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Sciences, Hunan Normal University, Changsha Hunan 410081, China
| | - Lu Wang
- The National & Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Sciences, Hunan Normal University, Changsha Hunan 410081, China
| | - Yi Zhuo
- The National & Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Sciences, Hunan Normal University, Changsha Hunan 410081, China; Hunan Provincical Key Laboratory of Neurorestoratology, the Second Affiliated Hospital of Hunan Normal University, Changsha Hunan, 410003, China
| | - Lang Long
- Hunan Provincical Key Laboratory of Neurorestoratology, the Second Affiliated Hospital of Hunan Normal University, Changsha Hunan, 410003, China
| | - Jiaomei Qi
- The National & Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Sciences, Hunan Normal University, Changsha Hunan 410081, China
| | - Li Hu
- Hunan Provincical Key Laboratory of Neurorestoratology, the Second Affiliated Hospital of Hunan Normal University, Changsha Hunan, 410003, China
| | - Da Duan
- Hunan Provincical Key Laboratory of Neurorestoratology, the Second Affiliated Hospital of Hunan Normal University, Changsha Hunan, 410003, China
| | - Ping Chen
- The National & Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Sciences, Hunan Normal University, Changsha Hunan 410081, China.
| | - Ming Lu
- The National & Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Sciences, Hunan Normal University, Changsha Hunan 410081, China; Hunan Provincical Key Laboratory of Neurorestoratology, the Second Affiliated Hospital of Hunan Normal University, Changsha Hunan, 410003, China.
| |
Collapse
|
19
|
Ye J, Wen Y, Chu X, Li P, Cheng B, Cheng S, Liu L, Zhang L, Ma M, Qi X, Liang C, Kafle OP, Jia Y, Wu C, Wang S, Wang X, Ning Y, Zhang F. Association between herpes simplex virus 1 exposure and the risk of depression in UK Biobank. Clin Transl Med 2020; 10:e108. [PMID: 32564518 PMCID: PMC7403656 DOI: 10.1002/ctm2.108] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2020] [Revised: 06/01/2020] [Accepted: 06/02/2020] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Herpes simplex virus-1 (HSV-1) infection is reported to be associated with depression. But limited efforts were made to investigate the relationship between HSV-1 infection and the risk of depression, especially from the genetic perspective. METHODS In UK Biobank cohort, linear and logistic regression analyses were first performed to test the association of HSV-1 seropositivity/antibody with depression, including depression status (N = 2951) and Patient Health Questionnaire (PHQ) score (N = 2839). Using individual genotypic and phenotypic data from the UK Biobank, genome-wide environmental interaction study (GWEIS) was then conducted by PLINK2.0 to evaluate gene × HSV-1 interacting effect on the risk of depression. Finally, gene set enrichment analysis was conducted to identify the biological pathways involved in the observed gene × HSV-1 interaction for depression. RESULT In UK Biobank cohort, significant associations were observed between depression status and HSV-1 (odds ratio [OR] = 1.09; 95% confidence interval [CI], 1.02-1.16; P = 2.40 × 10-2 for HSV-1 antibody and OR = 1.28; 95% CI, 1.12-1.47, P = 2.59 × 10-3 for HSV-1 seropositivity). GWEIS revealed four significant gene × HSV-1 interaction signals for PHQ score (all P < 5.0 × 10-8 ) and the leading loci was SULF2 (rs6094791, P = 8.60 × 10-9 ). Pathway analyses identified 21 pathways for PHQ score and 19 for depression status, including multiple neural development- and immune-related ones, such as KEGG_NEUROACTIVE_LIGAND_RECEPTOR_INTERACTION (false discovery rate [FDR] = 3.18 × 10-2 ) for depression and LU_AGING_BRAIN_UP (FDR = 4.21 × 10-2 ) for PHQ score. CONCLUSION Our results suggested that HSV-1 was associated with the risk of depression, which was modulated by the several genes that were related to the nerve development or immune function.
Collapse
Affiliation(s)
- Jing Ye
- Key Laboratory of Trace Elements and Endemic Diseases of National Health and Family Planning Commission, School of Public Health, Health Science Center, Xi'an Jiaotong University, Xi'an, China
| | - Yan Wen
- Key Laboratory of Trace Elements and Endemic Diseases of National Health and Family Planning Commission, School of Public Health, Health Science Center, Xi'an Jiaotong University, Xi'an, China
| | - Xiaomeng Chu
- Key Laboratory of Trace Elements and Endemic Diseases of National Health and Family Planning Commission, School of Public Health, Health Science Center, Xi'an Jiaotong University, Xi'an, China
| | - Ping Li
- Key Laboratory of Trace Elements and Endemic Diseases of National Health and Family Planning Commission, School of Public Health, Health Science Center, Xi'an Jiaotong University, Xi'an, China
| | - Bolun Cheng
- Key Laboratory of Trace Elements and Endemic Diseases of National Health and Family Planning Commission, School of Public Health, Health Science Center, Xi'an Jiaotong University, Xi'an, China
| | - Shiqiang Cheng
- Key Laboratory of Trace Elements and Endemic Diseases of National Health and Family Planning Commission, School of Public Health, Health Science Center, Xi'an Jiaotong University, Xi'an, China
| | - Li Liu
- Key Laboratory of Trace Elements and Endemic Diseases of National Health and Family Planning Commission, School of Public Health, Health Science Center, Xi'an Jiaotong University, Xi'an, China
| | - Lu Zhang
- Key Laboratory of Trace Elements and Endemic Diseases of National Health and Family Planning Commission, School of Public Health, Health Science Center, Xi'an Jiaotong University, Xi'an, China
| | - Mei Ma
- Key Laboratory of Trace Elements and Endemic Diseases of National Health and Family Planning Commission, School of Public Health, Health Science Center, Xi'an Jiaotong University, Xi'an, China
| | - Xin Qi
- Key Laboratory of Trace Elements and Endemic Diseases of National Health and Family Planning Commission, School of Public Health, Health Science Center, Xi'an Jiaotong University, Xi'an, China
| | - Chujun Liang
- Key Laboratory of Trace Elements and Endemic Diseases of National Health and Family Planning Commission, School of Public Health, Health Science Center, Xi'an Jiaotong University, Xi'an, China
| | - Om Prakash Kafle
- Key Laboratory of Trace Elements and Endemic Diseases of National Health and Family Planning Commission, School of Public Health, Health Science Center, Xi'an Jiaotong University, Xi'an, China
| | - Yumeng Jia
- Key Laboratory of Trace Elements and Endemic Diseases of National Health and Family Planning Commission, School of Public Health, Health Science Center, Xi'an Jiaotong University, Xi'an, China
| | - Cuiyan Wu
- Key Laboratory of Trace Elements and Endemic Diseases of National Health and Family Planning Commission, School of Public Health, Health Science Center, Xi'an Jiaotong University, Xi'an, China
| | - Sen Wang
- Key Laboratory of Trace Elements and Endemic Diseases of National Health and Family Planning Commission, School of Public Health, Health Science Center, Xi'an Jiaotong University, Xi'an, China
| | - Xi Wang
- Key Laboratory of Trace Elements and Endemic Diseases of National Health and Family Planning Commission, School of Public Health, Health Science Center, Xi'an Jiaotong University, Xi'an, China
| | - Yujie Ning
- Key Laboratory of Trace Elements and Endemic Diseases of National Health and Family Planning Commission, School of Public Health, Health Science Center, Xi'an Jiaotong University, Xi'an, China
| | - Feng Zhang
- Key Laboratory of Trace Elements and Endemic Diseases of National Health and Family Planning Commission, School of Public Health, Health Science Center, Xi'an Jiaotong University, Xi'an, China
| |
Collapse
|
20
|
Varderidou-Minasian S, Verheijen BM, Schätzle P, Hoogenraad CC, Pasterkamp RJ, Altelaar M. Deciphering the Proteome Dynamics during Development of Neurons Derived from Induced Pluripotent Stem Cells. J Proteome Res 2020; 19:2391-2403. [PMID: 32357013 PMCID: PMC7281779 DOI: 10.1021/acs.jproteome.0c00070] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
![]()
Neuronal development is a complex
multistep process that shapes
neurons by progressing though several typical stages, including axon
outgrowth, dendrite formation, and synaptogenesis. Knowledge of the
mechanisms of neuronal development is mostly derived from the study
of animal models. Advances in stem cell technology now enable us to
generate neurons from human induced pluripotent stem cells (iPSCs).
Here we provide a mass spectrometry-based quantitative proteomic signature
of human iPSC-derived neurons, i.e., iPSC-derived induced glutamatergic
neurons and iPSC-derived motor neurons, throughout neuronal differentiation.
Tandem mass tag 10-plex labeling was carried out to perform proteomic
profiling of cells at different time points. Our analysis reveals
significant expression changes (FDR < 0.001) of several key proteins
during the differentiation process, e.g., proteins involved in the
Wnt and Notch signaling pathways. Overall, our data provide a rich
resource of information on protein expression during human iPSC neuron
differentiation.
Collapse
Affiliation(s)
- Suzy Varderidou-Minasian
- Biomolecular Mass Spectrometry and Proteomics, Bijvoet Center for Biomolecular Research and Utrecht Institute for Pharmaceutical Sciences, University of Utrecht, Padualaan 8, 3584 CH Utrecht, The Netherlands.,Netherlands Proteomics Center, Padualaan 8, 3584 CH Utrecht, The Netherlands
| | - Bert M Verheijen
- Department of Translational Neuroscience, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht University, 3584 CG Utrecht, The Netherlands
| | - Philipp Schätzle
- Cell Biology, Department of Biology, Faculty of Science, Utrecht University, 3584 CH Utrecht, The Netherlands
| | - Casper C Hoogenraad
- Cell Biology, Department of Biology, Faculty of Science, Utrecht University, 3584 CH Utrecht, The Netherlands
| | - R Jeroen Pasterkamp
- Department of Translational Neuroscience, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht University, 3584 CG Utrecht, The Netherlands
| | - Maarten Altelaar
- Biomolecular Mass Spectrometry and Proteomics, Bijvoet Center for Biomolecular Research and Utrecht Institute for Pharmaceutical Sciences, University of Utrecht, Padualaan 8, 3584 CH Utrecht, The Netherlands.,Netherlands Proteomics Center, Padualaan 8, 3584 CH Utrecht, The Netherlands
| |
Collapse
|
21
|
Alonge KM, Logsdon AF, Murphree TA, Banks WA, Keene CD, Edgar JS, Whittington D, Schwartz MW, Guttman M. Quantitative analysis of chondroitin sulfate disaccharides from human and rodent fixed brain tissue by electrospray ionization-tandem mass spectrometry. Glycobiology 2020; 29:847-860. [PMID: 31361007 DOI: 10.1093/glycob/cwz060] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2019] [Revised: 07/25/2019] [Accepted: 07/25/2019] [Indexed: 02/07/2023] Open
Abstract
Chondroitin sulfates (CS) are long, negatively charged, unbranched glycosaminoglycan (GAG) chains attached to CS-proteoglycan (CSPG) core proteins that comprise the glycan component in both loose interstitial extracellular matrices (ECMs) and in rigid, structured perineuronal net (PNN) scaffolds within the brain. As aberrant CS-PNN formations have been linked to a range of pathological states, including Alzheimer's disease (AD) and schizophrenia, the analysis of CS-GAGs in brain tissue at the disaccharide level has great potential to enhance disease diagnosis and prognosis. Two mass-spectrometry (MS)-based approaches were adapted to detect CS disaccharides from minute fixed tissue samples with low picomolar sensitivity and high reproducibility. The first approach employed a straightforward, quantitative direct infusion (DI)-tandem mass spectrometry (MS/MS) technique to determine the percentages of Δ4S- and Δ6S-CS disaccharides within the 4S/6S-CS ratio, while the second used a comprehensive liquid chromatography (LC)-MS/MS technique to determine the relative percentages of Δ0S-, Δ4S-, Δ6S-, Δ4S6S-CS and Δ2S6S-CS disaccharides, with internal validation by full chondroitin lyase activity. The quantitative accuracy of the five primary biologically relevant CS disaccharides was validated using a developmental time course series in fixed rodent brain tissue. We then analyzed the CS disaccharide composition in formalin-fixed human brain tissue, thus providing the first quantitative report of CS sulfation patterns in the human brain. The ability to comprehensively analyze the CS disaccharide composition from fixed brain tissue provides a means with which to identify alterations in the CS-GAG composition in relation to the onset and/or progression of neurological diseases.
Collapse
Affiliation(s)
- Kimberly M Alonge
- University of Washington Medicine Diabetes Institute, Department of Medicine, Seattle, WA, USA
| | - Aric F Logsdon
- Department of Geriatric Research Education and Clinical Center (GRECC), Veterans Affairs Puget Sound Health Care System, Seattle, WA, USA.,Division of Gerontology and Geriatric Medicine, Department of Medicine, University of Washington, Seattle, WA, USA
| | - Taylor A Murphree
- Department of Medicinal Chemistry, University of Washington, Seattle, WA, USA
| | - William A Banks
- Department of Geriatric Research Education and Clinical Center (GRECC), Veterans Affairs Puget Sound Health Care System, Seattle, WA, USA.,Division of Gerontology and Geriatric Medicine, Department of Medicine, University of Washington, Seattle, WA, USA
| | - C Dirk Keene
- Division of Neuropathology, Department of Pathology, University of Washington, Seattle, WA, USA
| | - J Scott Edgar
- Department of Medicinal Chemistry, University of Washington, Seattle, WA, USA
| | - Dale Whittington
- Department of Medicinal Chemistry, University of Washington, Seattle, WA, USA
| | - Michael W Schwartz
- University of Washington Medicine Diabetes Institute, Department of Medicine, Seattle, WA, USA
| | - Miklos Guttman
- Department of Medicinal Chemistry, University of Washington, Seattle, WA, USA
| |
Collapse
|
22
|
Yoshida H, Koodie L, Jacobsen K, Hanzawa K, Miyamoto Y, Yamamoto M. B4GALNT1 induces angiogenesis, anchorage independence growth and motility, and promotes tumorigenesis in melanoma by induction of ganglioside GM2/GD2. Sci Rep 2020; 10:1199. [PMID: 31988291 PMCID: PMC6985110 DOI: 10.1038/s41598-019-57130-2] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2019] [Accepted: 11/22/2019] [Indexed: 12/11/2022] Open
Abstract
β-1,4-N-Acetyl-Galactosaminyltransferase 1 (B4GALNT1) encodes the key enzyme B4GALNT1 to generate gangliosides GM2/GD2. GM2/GD2 gangliosides are surface glycolipids mainly found on brain neurons as well as peripheral nerves and skin melanocytes and are reported to exacerbate the malignant potential of melanomas. In order to elucidate the mechanism, we performed functional analyses of B4GALNT1-overexpressing cells. We analyzed ganglioside pattern on four melanoma and two neuroblastoma cell lines by high performance liquid chromatography (HPLC). We overexpressed B4GALNT1 in GM2/GD2-negative human melanoma cell line (SH4) and confirmed production of GM2/GD2 by HPLC. They showed higher anchorage independence growth (AIG) in colony formation assay, and exhibited augmented motility. In vitro, cell proliferation was not affected by GM2/GD2 expression. In vivo, GM2/GD2-positive SH4 clones showed significantly higher tumorigenesis in NOD/Scid/IL2Rγ-null mice, and immunostaining of mouse CD31 revealed that GM2/GD2 induced remarkable angiogenesis. No differences were seen in melanoma stem cell and Epithelial-Mesenchymal Transition markers between GM2/GD2-positive and -negative SH4 cells. We therefore concluded that B4GALNT1, and consequently GM2/GD2, enhanced tumorigenesis via induction of angiogenesis, AIG, and cell motility. RNA-Seq suggested periostin as a potential key factor for angiogenesis and AIG. These findings may lead to development of novel therapy for refractory melanoma.
Collapse
Affiliation(s)
- Hideki Yoshida
- Department of Surgery, University of Minnesota, Minneapolis, Minnesota, USA
| | - Lisa Koodie
- Department of Surgery, University of Minnesota, Minneapolis, Minnesota, USA
| | - Kari Jacobsen
- Department of Surgery, University of Minnesota, Minneapolis, Minnesota, USA
| | - Ken Hanzawa
- Department of Molecular Biology, Osaka International Cancer Institute, Osaka, Japan
| | - Yasuhide Miyamoto
- Department of Molecular Biology, Osaka International Cancer Institute, Osaka, Japan
| | - Masato Yamamoto
- Department of Surgery, University of Minnesota, Minneapolis, Minnesota, USA.
- Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota, USA.
- Stem Cell Institute, University of Minnesota, Minneapolis, Minnesota, USA.
| |
Collapse
|
23
|
Korf-Klingebiel M, Reboll MR, Grote K, Schleiner H, Wang Y, Wu X, Klede S, Mikhed Y, Bauersachs J, Klintschar M, Rudat C, Kispert A, Niessen HW, Lübke T, Dierks T, Wollert KC. Heparan Sulfate-Editing Extracellular Sulfatases Enhance VEGF Bioavailability for Ischemic Heart Repair. Circ Res 2019; 125:787-801. [PMID: 31434553 DOI: 10.1161/circresaha.119.315023] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
RATIONALE Mechanistic insight into the inflammatory response after acute myocardial infarction may inform new molecularly targeted treatment strategies to prevent chronic heart failure. OBJECTIVE We identified the sulfatase SULF2 in an in silico secretome analysis in bone marrow cells from patients with acute myocardial infarction and detected increased sulfatase activity in myocardial autopsy samples. SULF2 (Sulf2 in mice) and its isoform SULF1 (Sulf1) act as endosulfatases removing 6-O-sulfate groups from heparan sulfate (HS) in the extracellular space, thus eliminating docking sites for HS-binding proteins. We hypothesized that the Sulfs have a role in tissue repair after myocardial infarction. METHODS AND RESULTS Both Sulfs were dynamically upregulated after coronary artery ligation in mice, attaining peak expression and activity levels during the first week after injury. Sulf2 was expressed by monocytes and macrophages, Sulf1 by endothelial cells and fibroblasts. Infarct border zone capillarization was impaired, scar size increased, and cardiac dysfunction more pronounced in mice with a genetic deletion of either Sulf1 or Sulf2. Studies in bone marrow-chimeric Sulf-deficient mice and Sulf-deficient cardiac endothelial cells established that inflammatory cell-derived Sulf2 and endothelial cell-autonomous Sulf1 promote angiogenesis. Mechanistically, both Sulfs reduced HS sulfation in the infarcted myocardium, thereby diminishing Vegfa (vascular endothelial growth factor A) interaction with HS. Along this line, both Sulfs rendered infarcted mouse heart explants responsive to the angiogenic effects of HS-binding Vegfa164 but did not modulate the angiogenic effects of non-HS-binding Vegfa120. Treating wild-type mice systemically with the small molecule HS-antagonist surfen (bis-2-methyl-4-amino-quinolyl-6-carbamide, 1 mg/kg/day) for 7 days after myocardial infarction released Vegfa from HS, enhanced infarct border-zone capillarization, and exerted sustained beneficial effects on cardiac function and survival. CONCLUSIONS These findings establish HS-editing Sulfs as critical inducers of postinfarction angiogenesis and identify HS sulfation as a therapeutic target for ischemic tissue repair.
Collapse
Affiliation(s)
- Mortimer Korf-Klingebiel
- From the Division of Molecular and Translational Cardiology (M.K.-K., M.R.R., K.G., H.S., Y.W., X.W., S.K., Y.M., K.C.W.), Hannover Medical School, Germany.,Department of Cardiology and Angiology (M.K.-K., M.R.R., K.G., H.S., Y.W., X.W., S.K., Y.M., J.B., K.C.W.), Hannover Medical School, Germany
| | - Marc R Reboll
- From the Division of Molecular and Translational Cardiology (M.K.-K., M.R.R., K.G., H.S., Y.W., X.W., S.K., Y.M., K.C.W.), Hannover Medical School, Germany.,Department of Cardiology and Angiology (M.K.-K., M.R.R., K.G., H.S., Y.W., X.W., S.K., Y.M., J.B., K.C.W.), Hannover Medical School, Germany
| | - Karsten Grote
- From the Division of Molecular and Translational Cardiology (M.K.-K., M.R.R., K.G., H.S., Y.W., X.W., S.K., Y.M., K.C.W.), Hannover Medical School, Germany.,Department of Cardiology and Angiology (M.K.-K., M.R.R., K.G., H.S., Y.W., X.W., S.K., Y.M., J.B., K.C.W.), Hannover Medical School, Germany
| | - Hauke Schleiner
- From the Division of Molecular and Translational Cardiology (M.K.-K., M.R.R., K.G., H.S., Y.W., X.W., S.K., Y.M., K.C.W.), Hannover Medical School, Germany.,Department of Cardiology and Angiology (M.K.-K., M.R.R., K.G., H.S., Y.W., X.W., S.K., Y.M., J.B., K.C.W.), Hannover Medical School, Germany
| | - Yong Wang
- From the Division of Molecular and Translational Cardiology (M.K.-K., M.R.R., K.G., H.S., Y.W., X.W., S.K., Y.M., K.C.W.), Hannover Medical School, Germany.,Department of Cardiology and Angiology (M.K.-K., M.R.R., K.G., H.S., Y.W., X.W., S.K., Y.M., J.B., K.C.W.), Hannover Medical School, Germany
| | - Xuekun Wu
- From the Division of Molecular and Translational Cardiology (M.K.-K., M.R.R., K.G., H.S., Y.W., X.W., S.K., Y.M., K.C.W.), Hannover Medical School, Germany.,Department of Cardiology and Angiology (M.K.-K., M.R.R., K.G., H.S., Y.W., X.W., S.K., Y.M., J.B., K.C.W.), Hannover Medical School, Germany
| | - Stefanie Klede
- From the Division of Molecular and Translational Cardiology (M.K.-K., M.R.R., K.G., H.S., Y.W., X.W., S.K., Y.M., K.C.W.), Hannover Medical School, Germany.,Department of Cardiology and Angiology (M.K.-K., M.R.R., K.G., H.S., Y.W., X.W., S.K., Y.M., J.B., K.C.W.), Hannover Medical School, Germany
| | - Yuliya Mikhed
- From the Division of Molecular and Translational Cardiology (M.K.-K., M.R.R., K.G., H.S., Y.W., X.W., S.K., Y.M., K.C.W.), Hannover Medical School, Germany.,Department of Cardiology and Angiology (M.K.-K., M.R.R., K.G., H.S., Y.W., X.W., S.K., Y.M., J.B., K.C.W.), Hannover Medical School, Germany
| | - Johann Bauersachs
- Department of Cardiology and Angiology (M.K.-K., M.R.R., K.G., H.S., Y.W., X.W., S.K., Y.M., J.B., K.C.W.), Hannover Medical School, Germany
| | | | - Carsten Rudat
- Institute of Molecular Biology (C.R., A.K.), Hannover Medical School, Germany
| | - Andreas Kispert
- Institute of Molecular Biology (C.R., A.K.), Hannover Medical School, Germany
| | - Hans W Niessen
- Department of Pathology and Department of Cardiac Surgery, Institute for Cardiovascular Research, VU University Medical Center, Amsterdam, The Netherlands (H.W.N.)
| | - Torben Lübke
- Department of Chemistry, Biochemistry I, Bielefeld University, Germany (T.L., T.D.)
| | - Thomas Dierks
- Department of Chemistry, Biochemistry I, Bielefeld University, Germany (T.L., T.D.)
| | - Kai C Wollert
- From the Division of Molecular and Translational Cardiology (M.K.-K., M.R.R., K.G., H.S., Y.W., X.W., S.K., Y.M., K.C.W.), Hannover Medical School, Germany.,Department of Cardiology and Angiology (M.K.-K., M.R.R., K.G., H.S., Y.W., X.W., S.K., Y.M., J.B., K.C.W.), Hannover Medical School, Germany
| |
Collapse
|
24
|
Heparan Sulfate Proteoglycan Synthesis Is Dysregulated in Human Osteoarthritic Cartilage. THE AMERICAN JOURNAL OF PATHOLOGY 2019; 189:632-647. [DOI: 10.1016/j.ajpath.2018.11.011] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/16/2018] [Revised: 11/06/2018] [Accepted: 11/13/2018] [Indexed: 12/11/2022]
|
25
|
|
26
|
Heparan sulfate S-domains and extracellular sulfatases (Sulfs): their possible roles in protein aggregation diseases. Glycoconj J 2018; 35:387-396. [PMID: 30003471 DOI: 10.1007/s10719-018-9833-8] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2018] [Revised: 06/25/2018] [Accepted: 07/04/2018] [Indexed: 10/28/2022]
Abstract
Highly sulfated domains of heparan sulfate (HS), also known as HS S-domains, consist of repeated trisulfated disaccharide units [iduronic acid (2S)-glucosamine (NS, 6S)-]. The expression of HS S-domains at the cell surface is determined by two mechanisms: tightly regulated biosynthetic machinery and enzymatic remodeling by extracellular endoglucosamine 6-sulfatases, Sulf-1 and Sulf-2. Intracellular or extracellular deposits of misfolded and aggregated proteins are characteristic of protein aggregation diseases. Although proteins can aggregate alone, deposits of protein aggregates in vivo contain a number of proteinaceous and non-protein components. HS S-domains are one non-protein component of these aggregated deposits. HS S-domains are considered to be critical for signal transduction of several growth factors and several disease conditions, such as tumor progression, but their roles in protein aggregation diseases are not yet fully understood. This review summarizes the current understanding of the possible roles of HS S-domains and Sulfs in the formation and cytotoxicity of protein aggregates.
Collapse
|
27
|
Abstract
Proteoglycans are diverse, complex extracellular/cell surface macromolecules composed of a central core protein with covalently linked glycosaminoglycan (GAG) chains; both of these components contribute to the growing list of important bio-active functions attributed to proteoglycans. Increasingly, attention has been paid to the roles of proteoglycans in nervous tissue development due to their highly regulated spatio/temporal expression patterns, whereby they promote/inhibit neurite outgrowth, participate in specification and maturation of various precursor cell types, and regulate cell behaviors like migration, axonal pathfinding, synaptogenesis and plasticity. These functions emanate from both the environments proteoglycans create around cells by retaining ions and water or serving as scaffolds for cell shaping or motility, and from dynamic interactions that modulate signaling fields for cytokines, growth factors and morphogens, which may bind to either the protein or GAG portions. Also, genetic abnormalities impacting proteoglycan synthesis during critical steps of brain development and response to environmental insults and injuries, as well as changes in microenvironment interactions leading to tumors in the central nervous system, all suggest roles for proteoglycans in behavioral and intellectual disorders and malignancies.
Collapse
Affiliation(s)
- Nancy B Schwartz
- Department of Pediatrics, Biological Sciences Division, The University of Chicago, IL, USA.,Department of Biochemistry and Molecular Biology, Biological Sciences Division, The University of Chicago, IL, USA
| | - Miriam S Domowicz
- Department of Pediatrics, Biological Sciences Division, The University of Chicago, IL, USA
| |
Collapse
|
28
|
Lou X, Sun B, Song J, Wang Y, Jiang J, Xu Y, Ren Z, Su C. Human sulfatase 1 exerts anti-tumor activity by inhibiting the AKT/ CDK4 signaling pathway in melanoma. Oncotarget 2018; 7:84486-84495. [PMID: 27806323 PMCID: PMC5356675 DOI: 10.18632/oncotarget.12996] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2016] [Accepted: 10/24/2016] [Indexed: 01/22/2023] Open
Abstract
Human sulfatase 1 (hSulf-1) has aryl sulfatase activity. It can reduce the sulfation of cell surface heparan sulfate proteoglycan (HSPG) and inhibit various growth factor receptor-mediated signaling pathways. In most cancers, hSulf-1 is inactivated, which endows cancer cells with increasesed cell proliferation and metastatic activities, inhibition of apoptosis, and decreased sensitivity to radio- and chemotherapy. In this study, we found that hSulf-1 overexpression in melanoma cells can inhibit cell proliferation and induce cell cycle arrest and apoptosis by decreasing the protein kinase B (AKT) phosphorylation and limiting CDK4 nuclear import. We further confirmed that hSulf-1 overexpression can inhibit AKT phosphorylation and CDK4 nuclear localization and retard the growth of melanoma xenograft tumors in nude mice. Overall, hSulf-1 function in melanoma cells provides an ideal molecular treatment target. An important anti-tumor mechanism of hSulf-1 operates by decreasing downstream AKT signaling pathway activity and inhibiting the nuclear import of CDK4.
Collapse
Affiliation(s)
- Xiaoli Lou
- Department of Reconstructive Surgery, Changhai Hospital, Second Military Medical University, Shanghai 200433, China
| | - Bin Sun
- Department of Molecular Oncology, Eastern Hepatobiliary Surgery Hospital and National Center of Liver Cancer, Second Military Medical University, Shanghai 200433, China
| | - Jianxing Song
- Department of Reconstructive Surgery, Changhai Hospital, Second Military Medical University, Shanghai 200433, China
| | - Yicun Wang
- Department of Orthopedics, Jinling Hospital, Nanjing University Medical College, Nanjing 210002, China
| | - Junhao Jiang
- Cancer Center for Collaborative Innovation, Affiliated Peixian People's Hospital of Xuzhou Medical University, Jiangsu Peixian 221600, China
| | - Yang Xu
- Department of Molecular Oncology, Eastern Hepatobiliary Surgery Hospital and National Center of Liver Cancer, Second Military Medical University, Shanghai 200433, China
| | - Zeqiang Ren
- Cancer Center for Collaborative Innovation, Affiliated Peixian People's Hospital of Xuzhou Medical University, Jiangsu Peixian 221600, China
| | - Changqing Su
- Department of Molecular Oncology, Eastern Hepatobiliary Surgery Hospital and National Center of Liver Cancer, Second Military Medical University, Shanghai 200433, China.,Cancer Center for Collaborative Innovation, Affiliated Peixian People's Hospital of Xuzhou Medical University, Jiangsu Peixian 221600, China
| |
Collapse
|
29
|
Saied-Santiago K, Bülow HE. Diverse roles for glycosaminoglycans in neural patterning. Dev Dyn 2018; 247:54-74. [PMID: 28736980 PMCID: PMC5866094 DOI: 10.1002/dvdy.24555] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2017] [Revised: 07/13/2017] [Accepted: 07/17/2017] [Indexed: 01/11/2023] Open
Abstract
The nervous system coordinates the functions of most multicellular organisms and their response to the surrounding environment. Its development involves concerted cellular interactions, including migration, axon guidance, and synapse formation. These processes depend on the molecular constituents and structure of the extracellular matrices (ECM). An essential component of ECMs are proteoglycans, i.e., proteins containing unbranched glycan chains known as glycosaminoglycans (GAGs). A defining characteristic of GAGs is their enormous molecular diversity, created by extensive modifications of the glycans during their biosynthesis. GAGs are widely expressed, and their loss can lead to catastrophic neuronal defects. Despite their importance, we are just beginning to understand the function and mechanisms of GAGs in neuronal development. In this review, we discuss recent evidence suggesting GAGs have specific roles in neuronal patterning and synaptogenesis. We examine the function played by the complex modifications present on GAG glycans and their roles in regulating different aspects of neuronal patterning. Moreover, the review considers the function of proteoglycan core proteins in these processes, stressing their likely role as co-receptors of different signaling pathways in a redundant and context-dependent manner. We conclude by discussing challenges and future directions toward a better understanding of these fascinating molecules during neuronal development. Developmental Dynamics 247:54-74, 2018. © 2017 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
| | - Hannes E. Bülow
- Department of Genetics, Albert Einstein College of Medicine, Bronx, New York, 10461
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, New York, 10461
| |
Collapse
|
30
|
Yu P, Pearson CS, Geller HM. Flexible Roles for Proteoglycan Sulfation and Receptor Signaling. Trends Neurosci 2018; 41:47-61. [PMID: 29150096 PMCID: PMC5748001 DOI: 10.1016/j.tins.2017.10.005] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2017] [Revised: 10/19/2017] [Accepted: 10/25/2017] [Indexed: 11/25/2022]
Abstract
Proteoglycans (PGs) in the extracellular matrix (ECM) play vital roles in axon growth and navigation, plasticity, and regeneration of injured neurons. Different classes of PGs may support or inhibit cell growth, and their functions are determined in part by highly specific structural features. Among these, the pattern of sulfation on the PG sugar chains is a paramount determinant of a diverse and flexible set of outcomes. Recent studies of PG sulfation illustrate the challenges of attributing biological actions to specific sulfation patterns, and suggest ways in which highly similar molecules may exert opposing effects on neurons. The receptors for PGs, which have yet to be fully characterized, display a similarly nuanced spectrum of effects. Different classes of PG function via overlapping families of receptors and signaling pathways. This enables them to control axon growth and guidance with remarkable specificity, but it poses challenges for determining the precise binding interactions and downstream effects of different PGs and their assorted sulfated epitopes. This review examines existing and emerging evidence for the roles of PG sulfation and receptor interactions in determining how these complex molecules influence neuronal development, growth, and function.
Collapse
Affiliation(s)
- Panpan Yu
- Guangdong-Hongkong-Macau Institute of CNS Regeneration; Ministry of Education Joint International Research Laboratory of CNS Regeneration, Jinan University, Guangzhou 510632, China.
| | - Craig S Pearson
- Laboratory of Developmental Neurobiology, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA; Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
| | - Herbert M Geller
- Laboratory of Developmental Neurobiology, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
31
|
Okada T, Keino-Masu K, Nagamine S, Kametani F, Ohto T, Hasegawa M, van Kuppevelt TH, Kunita S, Takahashi S, Masu M. Desulfation of Heparan Sulfate by Sulf1 and Sulf2 Is Required for Corticospinal Tract Formation. Sci Rep 2017; 7:13847. [PMID: 29062064 PMCID: PMC5653861 DOI: 10.1038/s41598-017-14185-3] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2017] [Accepted: 10/06/2017] [Indexed: 12/31/2022] Open
Abstract
Heparan sulfate (HS) has been implicated in a wide range of cell signaling. Here we report a novel mechanism in which extracellular removal of 6-O-sulfate groups from HS by the endosulfatases, Sulf1 and Sulf2, is essential for axon guidance during development. In Sulf1/2 double knockout (DKO) mice, the corticospinal tract (CST) was dorsally displaced on the midbrain surface. In utero electroporation of Sulf1/2 into radial glial cells along the third ventricle, where Sulf1/2 mRNAs are normally expressed, rescued the CST defects in the DKO mice. Proteomic analysis and functional testing identified Slit2 as the key molecule associated with the DKO phenotype. In the DKO brain, 6-O-sulfated HS was increased, leading to abnormal accumulation of Slit2 protein on the pial surface of the cerebral peduncle and hypothalamus, which caused dorsal repulsion of CST axons. Our findings indicate that postbiosynthetic desulfation of HS by Sulfs controls CST axon guidance through fine-tuning of Slit2 presentation.
Collapse
Affiliation(s)
- Takuya Okada
- Department of Molecular Neurobiology, Faculty of Medicine, University of Tsukuba, 1-1-1 Tennodai, Ibaraki, 305-8575, Japan
| | - Kazuko Keino-Masu
- Department of Molecular Neurobiology, Faculty of Medicine, University of Tsukuba, 1-1-1 Tennodai, Ibaraki, 305-8575, Japan
| | - Satoshi Nagamine
- Department of Molecular Neurobiology, Faculty of Medicine, University of Tsukuba, 1-1-1 Tennodai, Ibaraki, 305-8575, Japan.,Pharmaceuticals and Medical Devices Agency, 3-3-2 Kasumigaseki, Chiyoda-Ku, Tokyo, 100-0013, Japan
| | - Fuyuki Kametani
- Department of Neuropathology and Cell Biology, Tokyo Metropolitan Institute of Medical Science, Setagaya-ku, Tokyo, 156-8506, Japan
| | - Tatsuyuki Ohto
- Department of Molecular Neurobiology, Faculty of Medicine, University of Tsukuba, 1-1-1 Tennodai, Ibaraki, 305-8575, Japan.,Department of Pediatrics, University of Tsukuba Hospital, 2-1-1 Amakubo, Ibaraki, 305-8576, Japan
| | - Masato Hasegawa
- Department of Neuropathology and Cell Biology, Tokyo Metropolitan Institute of Medical Science, Setagaya-ku, Tokyo, 156-8506, Japan
| | - Toin H van Kuppevelt
- Department of Biochemistry, Nijmegen Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Satoshi Kunita
- Laboratory Animal Resource Center, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki, 305-8575, Japan.,Center for Experimental Medicine, Jichi Medical University, 3311-1 Yakushiji, Shimotsuke, Tochigi, 329-0498, Japan
| | - Satoru Takahashi
- Laboratory Animal Resource Center, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki, 305-8575, Japan
| | - Masayuki Masu
- Department of Molecular Neurobiology, Faculty of Medicine, University of Tsukuba, 1-1-1 Tennodai, Ibaraki, 305-8575, Japan.
| |
Collapse
|
32
|
Yu C, Griffiths LR, Haupt LM. Exploiting Heparan Sulfate Proteoglycans in Human Neurogenesis-Controlling Lineage Specification and Fate. Front Integr Neurosci 2017; 11:28. [PMID: 29089873 PMCID: PMC5650988 DOI: 10.3389/fnint.2017.00028] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2017] [Accepted: 09/25/2017] [Indexed: 12/26/2022] Open
Abstract
Unspecialized, self-renewing stem cells have extraordinary application to regenerative medicine due to their multilineage differentiation potential. Stem cell therapies through replenishing damaged or lost cells in the injured area is an attractive treatment of brain trauma and neurodegenerative neurological disorders. Several stem cell types have neurogenic potential including neural stem cells (NSCs), embryonic stem cells (ESCs), induced pluripotent stem cells (iPSCs), and mesenchymal stem cells (MSCs). Currently, effective use of these cells is limited by our lack of understanding and ability to direct lineage commitment and differentiation of neural lineages. Heparan sulfate proteoglycans (HSPGs) are ubiquitous proteins within the stem cell microenvironment or niche and are found localized on the cell surface and in the extracellular matrix (ECM), where they interact with numerous signaling molecules. The glycosaminoglycan (GAG) chains carried by HSPGs are heterogeneous carbohydrates comprised of repeating disaccharides with specific sulfation patterns that govern ligand interactions to numerous factors including the fibroblast growth factors (FGFs) and wingless-type MMTV integration site family (Wnts). As such, HSPGs are plausible targets for guiding and controlling neural stem cell lineage fate. In this review, we provide an overview of HSPG family members syndecans and glypicans, and perlecan and their role in neurogenesis. We summarize the structural changes and subsequent functional implications of heparan sulfate as cells undergo neural lineage differentiation as well as outline the role of HSPG core protein expression throughout mammalian neural development and their function as cell receptors and co-receptors. Finally, we highlight suitable biomimetic approaches for exploiting the role of HSPGs in mammalian neurogenesis to control and tailor cell differentiation into specific lineages. An improved ability to control stem cell specific neural lineage fate and produce abundant cells of lineage specificity will further advance stem cell therapy for the development of improved repair of neurological disorders. We propose a deeper understanding of HSPG-mediated neurogenesis can potentially provide novel therapeutic targets of neurogenesis.
Collapse
Affiliation(s)
- Chieh Yu
- Genomics Research Centre, Institute of Health and Biomedical Innovation, School of Biomedical Sciences, Queensland University of Technology, Brisbane, QLD, Australia
| | - Lyn R Griffiths
- Genomics Research Centre, Institute of Health and Biomedical Innovation, School of Biomedical Sciences, Queensland University of Technology, Brisbane, QLD, Australia
| | - Larisa M Haupt
- Genomics Research Centre, Institute of Health and Biomedical Innovation, School of Biomedical Sciences, Queensland University of Technology, Brisbane, QLD, Australia
| |
Collapse
|
33
|
Roberts RO, Kang YN, Hu C, Moser CD, Wang S, Moore MJ, Graham RP, Lai JP, Petersen RC, Roberts LR. Decreased Expression of Sulfatase 2 in the Brains of Alzheimer's Disease Patients: Implications for Regulation of Neuronal Cell Signaling. J Alzheimers Dis Rep 2017; 1:115-124. [PMID: 30035253 PMCID: PMC6052874 DOI: 10.3233/adr-170028] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
Background: The human sulfatase 1 (SULF1) and sulfatase 2 (SULF2) genes modulate cell signaling and homeostasis in many tissues. Gene expression analyses have implicated SULF2 in disease pathogenesis, including Alzheimer’s disease (AD), but changes in brain SULF2 expression have not been directly established. Objective: To investigate the expression of SULF1 and SULF2 in brain tissues from AD cases and cognitively normal controls. Methods: Autopsy tissue from AD cases (n = 20) and age-and gender-matched cognitively normal controls (n = 20) were identified from the Mayo Clinic Alzheimer’s Disease Patient Registry neuropathology database. Tissue slides were stained for SULF1 and SULF2 protein expression in the hippocampus and frontal lobe and an expression score computed from the proportion of cells stained and the intensity of staining (range 0 [no expression] to 9 [marked expression]). Results: SULF2 expression was reduced in AD cases. Compared to cognitively normal controls, SULF2 expression in AD cases was significantly decreased in the hippocampal Cornu Ammonis (CA) (mean score of 6.5 in cases versus 8.3 in controls; p = 0.003), in the gray matter of the parahippocampal gyrus (5.6 in cases versus 7.6 in controls; p = 0.003), and in the frontal lobe gray matter (5.4 in cases versus 7.4 in controls; p = 0.002). There was no difference in SULF1 expression in the hippocampus or frontal lobe of AD cases and controls. As expected there were no differences in SULF1 or SULF2 expression in white matter in AD cases compared to cognitively normal controls. Conclusion: Decreased SULF2 in specific regions of the brain occurs in AD.
Collapse
Affiliation(s)
- Rosebud O Roberts
- Department of Health Sciences Research, Mayo Clinic, Rochester, MN, USA.,Department of Neurology, Mayo Clinic, Rochester, MN, USA
| | - Yoo Na Kang
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN, USA.,Department of Pathology, Keimyung University, Daegu, South Korea
| | - Chunling Hu
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN, USA
| | - Catherine D Moser
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN, USA
| | - Shaoqing Wang
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN, USA
| | - Michael J Moore
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN, USA
| | - Rondell P Graham
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, USA
| | - Jin-Ping Lai
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN, USA
| | - Ronald C Petersen
- Department of Health Sciences Research, Mayo Clinic, Rochester, MN, USA.,Department of Neurology, Mayo Clinic, Rochester, MN, USA
| | - Lewis R Roberts
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN, USA
| |
Collapse
|
34
|
Changyaleket B, Deliu Z, Chignalia AZ, Feinstein DL. Heparanase: Potential roles in multiple sclerosis. J Neuroimmunol 2017; 310:72-81. [PMID: 28778449 DOI: 10.1016/j.jneuroim.2017.07.001] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2017] [Revised: 06/22/2017] [Accepted: 07/01/2017] [Indexed: 12/14/2022]
Abstract
Heparanase is a heparan sulfate degrading enzyme that cleaves heparan sulfate (HS) chains present on HS proteoglycans (HSPGs), and has been well characterized for its roles in tumor metastasis and inflammation. However, heparanase is emerging as a contributing factor in the genesis and severity of a variety of neurodegenerative diseases and conditions. This is in part due to the wide variety of HSPGs on which the presence or absence of HS moieties dictates protein function. This includes growth factors, chemokines, cytokines, as well as components of the extracellular matrix (ECM) which in turn regulate leukocyte infiltration into the CNS. Roles for heparanase in stroke, Alzheimer's disease, and glioma growth have been described; roles for heparanase in other disease such as multiple sclerosis (MS) are less well established. However, given its known roles in inflammation and leukocyte infiltration, it is likely that heparanase also contributes to MS pathology. In this review, we will briefly summarize what is known about heparanase roles in the CNS, and speculate as to its potential role in regulating disease progression in MS and its animal model EAE (experimental autoimmune encephalitis), which may justify testing of heparanase inhibitors for MS treatment.
Collapse
Affiliation(s)
| | - Zane Deliu
- Department of Anesthesiology, University of Illinois, Chicago, IL 60612, USA
| | - Andreia Z Chignalia
- Department of Anesthesiology, University of Illinois, Chicago, IL 60612, USA
| | - Douglas L Feinstein
- Department of Anesthesiology, University of Illinois, Chicago, IL 60612, USA; Jesse Brown Veteran Affairs Medical Center, Chicago, IL 60612, USA.
| |
Collapse
|
35
|
The "in and out" of glucosamine 6-O-sulfation: the 6th sense of heparan sulfate. Glycoconj J 2016; 34:285-298. [PMID: 27812771 DOI: 10.1007/s10719-016-9736-5] [Citation(s) in RCA: 59] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2016] [Revised: 09/26/2016] [Accepted: 09/28/2016] [Indexed: 01/06/2023]
Abstract
The biological properties of Heparan sulfate (HS) polysaccharides essentially rely on their ability to bind and modulate a multitude of protein ligands. These interactions involve internal oligosaccharide sequences defined by their sulfation patterns. Amongst these, the 6-O-sulfation of HS contributes significantly to the polysaccharide structural diversity and is critically involved in the binding of many proteins. HS 6-O-sulfation is catalyzed by 6-O-sulfotransferases (6OSTs) during biosynthesis, and it is further modified by the post-synthetic action of 6-O-endosulfatases (Sulfs), two enzyme families that remain poorly characterized. The aim of the present review is to summarize the contribution of 6-O-sulfates in HS structure/function relationships and to discuss the present knowledge on the complex mechanisms regulating HS 6-O-sulfation.
Collapse
|
36
|
Yi B, Qiu Y, Ji W, Wei M, Liu C, Peng Z, Zhang Y, Quan Z, Tang Z, Su C. Desulfation of cell surface HSPG is an effective strategy for the treatment of gallbladder carcinoma. Cancer Lett 2016; 381:349-58. [DOI: 10.1016/j.canlet.2016.08.002] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2016] [Revised: 07/30/2016] [Accepted: 08/02/2016] [Indexed: 01/08/2023]
|
37
|
O'Neill P, Lindsay SL, Pantiru A, Guimond SE, Fagoe N, Verhaagen J, Turnbull JE, Riddell JS, Barnett SC. Sulfatase-mediated manipulation of the astrocyte-Schwann cell interface. Glia 2016; 65:19-33. [PMID: 27535874 PMCID: PMC5244676 DOI: 10.1002/glia.23047] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2016] [Revised: 07/29/2016] [Accepted: 08/01/2016] [Indexed: 12/29/2022]
Abstract
Schwann cell (SC) transplantation following spinal cord injury (SCI) may have therapeutic potential. Functional recovery is limited however, due to poor SC interactions with host astrocytes and the induction of astrogliosis. Olfactory ensheathing cells (OECs) are closely related to SCs, but intermix more readily with astrocytes in culture and induce less astrogliosis. We previously demonstrated that OECs express higher levels of sulfatases, enzymes that remove 6-O-sulfate groups from heparan sulphate proteoglycans, than SCs and that RNAi knockdown of sulfatase prevented OEC-astrocyte mixing in vitro. As human OECs are difficult to culture in large numbers we have genetically engineered SCs using lentiviral vectors to express sulfatase 1 and 2 (SC-S1S2) and assessed their ability to interact with astrocytes. We demonstrate that SC-S1S2s have increased integrin-dependent motility in the presence of astrocytes via modulation of NRG and FGF receptor-linked PI3K/AKT intracellular signaling and do not form boundaries with astrocytes in culture. SC-astrocyte mixing is dependent on local NRG concentration and we propose that sulfatase enzymes influence the bioavailability of NRG ligand and thus influence SC behavior. We further demonstrate that injection of sulfatase expressing SCs into spinal cord white matter results in less glial reactivity than control SC injections comparable to that of OEC injections. Our data indicate that sulfatase-mediated modification of the extracellular matrix can influence glial interactions with astrocytes, and that SCs engineered to express sulfatase may be more OEC-like in character. This approach may be beneficial for cell transplant-mediated spinal cord repair. GLIA 2016 GLIA 2017;65:19-33.
Collapse
Affiliation(s)
- Paul O'Neill
- Institute of Infection, Inflammation and Immunity, 120 University Place, University of Glasgow, Glasgow, G12 8TA, United Kingdom
| | - Susan L Lindsay
- Institute of Infection, Inflammation and Immunity, 120 University Place, University of Glasgow, Glasgow, G12 8TA, United Kingdom
| | - Andreea Pantiru
- Institute of Infection, Inflammation and Immunity, 120 University Place, University of Glasgow, Glasgow, G12 8TA, United Kingdom
| | - Scott E Guimond
- Department of Biochemistry, Centre for Glycobiology, Institute of Integrative Biology, University of Liverpool, Liverpool, L69 7ZB, United Kingdom
| | - Nitish Fagoe
- Laboratory for Neuroregeneration, Netherlands Institute for Neuroscience, Meibergdreef 47, Amsterdam, BA, 1105, the Netherlands
| | - Joost Verhaagen
- Laboratory for Neuroregeneration, Netherlands Institute for Neuroscience, Meibergdreef 47, Amsterdam, BA, 1105, the Netherlands
| | - Jeremy E Turnbull
- Department of Biochemistry, Centre for Glycobiology, Institute of Integrative Biology, University of Liverpool, Liverpool, L69 7ZB, United Kingdom
| | - John S Riddell
- Institute of Neuroscience and Psychology, West Medical Building, University of Glasgow, Glasgow, G12 8QQ, United Kingdom
| | - Susan C Barnett
- Institute of Infection, Inflammation and Immunity, 120 University Place, University of Glasgow, Glasgow, G12 8TA, United Kingdom
| |
Collapse
|
38
|
Coulson-Thomas VJ. The role of heparan sulphate in development: the ectodermal story. Int J Exp Pathol 2016; 97:213-29. [PMID: 27385054 DOI: 10.1111/iep.12180] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2015] [Accepted: 02/24/2016] [Indexed: 12/27/2022] Open
Abstract
Heparan sulphate (HS) is ubiquitously expressed and is formed of repeating glucosamine and glucuronic/iduronic acid units which are generally highly sulphated. HS is found in tissues bound to proteins forming HS proteoglycans (HSPGs) which are present on the cell membrane or in the extracellular matrix. HSPGs influence a variety of biological processes by interacting with physiologically important proteins, such as morphogens, creating storage pools, generating morphogen gradients and directly mediating signalling pathways, thereby playing vital roles during development. This review discusses the vital role HS plays in the development of tissues from the ectodermal lineage. The ectodermal layer differentiates to form the nervous system (including the spine, peripheral nerves and brain), eye, epidermis, skin appendages and tooth enamel.
Collapse
|